Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Biomol Struct Dyn ; : 1-19, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37655680

ABSTRACT

Decaprenylphosphoryl-ß-d-ribose-2'-epimerase (DprE1) is a druggable target which is being exploited for the development of new anti-TB agents. In the present work, we report developing a pharmacophore model and performing virtual screening of Asinex database using the developed pharmacophore model to get eight hits as potential DprE1 inhibitors. The hits were used as leads to design new 3-phenylpyrazolo[1,5-a]pyrimidine-2,7(1H,4H)-dione based potential anti-TB agents. On the basis of the identified lead molecules, a total of 18 compounds were synthesized and evaluated for their anti-TB activity by using MABA. ADMET predictions for all the compounds revealed that these compounds have drug-like and lead-like properties. One of the final compounds was found to exhibit potent anti-TB activity against Mycobacterium bovis.Communicated by Ramaswamy H. Sarma.

2.
Hum Mol Genet ; 32(1): 30-45, 2023 01 01.
Article in English | MEDLINE | ID: mdl-35908190

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by an inherited unstable HTT CAG repeat that expands further, thereby eliciting a disease process that may be initiated by polyglutamine-expanded huntingtin or a short polyglutamine-product. Phosphorylation of selected candidate residues is reported to mediate polyglutamine-fragment degradation and toxicity. Here to support the discovery of phosphosites involved in the life-cycle of (full-length) huntingtin, we employed mass spectrometry-based phosphoproteomics to systematically identify sites in purified huntingtin and in the endogenous protein by proteomic and phosphoproteomic analyses of members of an HD neuronal progenitor cell panel. Our results bring total huntingtin phosphosites to 95, with more located in the N-HEAT domain relative to numbers in the Bridge and C-HEAT domains. Moreover, phosphorylation of C-HEAT Ser2550 by cAMP-dependent protein kinase (PKA), the top hit in kinase activity screens, was found to hasten huntingtin degradation, such that levels of the catalytic subunit (PRKACA) were inversely related to huntingtin levels. Taken together, these findings highlight categories of phosphosites that merit further study and provide a phosphosite kinase pair (pSer2550-PKA) with which to investigate the biological processes that regulate huntingtin degradation and thereby influence the steady state levels of huntingtin in HD cells.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Huntington Disease , Humans , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Hot Temperature , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Phosphorylation , Protein Domains , Proteomics
3.
Structure ; 28(9): 1035-1050.e8, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32668197

ABSTRACT

The polyQ expansion in huntingtin protein (HTT) is the prime cause of Huntington's disease (HD). The recent cryoelectron microscopy (cryo-EM) structure of HTT-HAP40 complex provided the structural information on its HEAT-repeat domains. Here, we present analyses of the impact of polyQ length on the structure and function of HTT via an integrative structural and biochemical approach. The cryo-EM analysis of normal (Q23) and disease (Q78) type HTTs shows that the structures of apo HTTs significantly differ from the structure of HTT in a HAP40 complex and that the polyQ expansion induces global structural changes in the relative movements among the HTT domains. In addition, we show that the polyQ expansion alters the phosphorylation pattern across HTT and that Ser2116 phosphorylation in turn affects the global structure and function of HTT. These results provide a molecular basis for the effect of the polyQ segment on HTT structure and activity, which may be important for HTT pathology.


Subject(s)
Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Peptides/metabolism , Cryoelectron Microscopy , Humans , Huntingtin Protein/genetics , Hydrogen Deuterium Exchange-Mass Spectrometry , Mass Spectrometry , Models, Molecular , Mutation , Peptides/chemistry , Phosphorylation , Protein Domains , Scattering, Small Angle , Serine/metabolism , X-Ray Diffraction
4.
PLoS Genet ; 15(3): e1007765, 2019 03.
Article in English | MEDLINE | ID: mdl-30897080

ABSTRACT

Rare individuals with inactivating mutations in the Huntington's disease gene (HTT) exhibit variable abnormalities that imply essential HTT roles during organ development. Here we report phenotypes produced when increasingly severe hypomorphic mutations in the murine HTT orthologue Htt, (HdhneoQ20, HdhneoQ50, HdhneoQ111), were placed over a null allele (Hdhex4/5). The most severe hypomorphic allele failed to rescue null lethality at gastrulation, while the intermediate, though still severe, alleles yielded recessive perinatal lethality and a variety of fetal abnormalities affecting body size, skin, skeletal and ear formation, and transient defects in hematopoiesis. Comparative molecular analysis of wild-type and Htt-null retinoic acid-differentiated cells revealed gene network dysregulation associated with organ development that nominate polycomb repressive complexes and miRNAs as molecular mediators. Together these findings demonstrate that Htt is required both pre- and post-gastrulation to support normal development.


Subject(s)
Huntingtin Protein/genetics , Huntington Disease/genetics , Alleles , Animals , Cell Differentiation/genetics , Disease Models, Animal , Gene Frequency/genetics , Genotype , Huntingtin Protein/physiology , Mice/embryology , Mutation , Nerve Tissue Proteins/genetics , Phenotype
5.
eNeuro ; 5(4)2018.
Article in English | MEDLINE | ID: mdl-30310861

ABSTRACT

The neurodegenerative Huntington's disease (HD) is caused by a polyglutamine (polyQ) amplification in the huntingtin protein (HTT). Currently there is no effective therapy available for HD; however, several efforts are directed to develop and optimize HTT-lowering methods to improve HD phenotypes. To validate these approaches, there is an immediate need for reliable, sensitive, and easily accessible methods to quantify HTT expression. Using the AlphaLISA platform, we developed two novel sensitive and robust assays for quantification of HTT in biological samples using commercially available antibodies. The first, a polyQ-independent assay, measures the total pool of HTT, while the second, a polyQ-dependent assay, preferentially detects the mutant form of HTT. Using purified HTT protein standards and brain homogenates from an HD mouse model, we determine a lower limit of quantification of 1 and 3 pm and optimal reproducibility with CV values lower than 7% for intra- and 20% for interassay. In addition, we used the assays to quantify HTT in neural stem cells generated from patient-derived induced pluripotent stem cells in vitro and in human brain tissue lysates. Finally, we could detect changes in HTT levels in a mouse model where mutant HTT was conditionally deleted in neural tissue, verifying the potential to monitor the outcome of HTT-lowering strategies. This analytical platform is ideal for high-throughput screens and thus has an added value for the HD community as a tool to optimize novel therapeutic approaches aimed at modulating HTT protein levels.


Subject(s)
Huntingtin Protein/analysis , Huntington Disease/diagnosis , Immunoassay/standards , Animals , Disease Models, Animal , HEK293 Cells , Humans , Immunoassay/methods , Mice , Mutation , Neural Stem Cells , Reproducibility of Results
6.
Mol Ther Nucleic Acids ; 11: 416-428, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29858077

ABSTRACT

The CAG repeat expansion that elongates the polyglutamine tract in huntingtin is the root genetic cause of Huntington's disease (HD), a debilitating neurodegenerative disorder. This seemingly slight change to the primary amino acid sequence alters the physical structure of the mutant protein and alters its activity. We have identified a set of G-quadruplex-forming DNA aptamers (MS1, MS2, MS3, MS4) that bind mutant huntingtin proximal to lysines K2932/K2934 in the C-terminal CTD-II domain. Aptamer binding to mutant huntingtin abrogated the enhanced polycomb repressive complex 2 (PRC2) stimulatory activity conferred by the expanded polyglutamine tract. In HD, but not normal, neuronal progenitor cells (NPCs), MS3 aptamer co-localized with endogenous mutant huntingtin and was associated with significantly decreased PRC2 activity. Furthermore, MS3 transfection protected HD NPCs against starvation-dependent stress with increased ATP. Therefore, DNA aptamers can preferentially target mutant huntingtin and modulate a gain of function endowed by the elongated polyglutamine segment. These mutant huntingtin binding aptamers provide novel molecular tools for delineating the effects of the HD mutation and encourage mutant huntingtin structure-based approaches to therapeutic development.

7.
J Proteome Res ; 16(8): 2692-2708, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28653853

ABSTRACT

Post-translational modifications (PTMs) of proteins regulate various cellular processes. PTMs of polyglutamine-expanded huntingtin (Htt) protein, which causes Huntington's disease (HD), are likely modulators of HD pathogenesis. Previous studies have identified and characterized several PTMs on exogenously expressed Htt fragments, but none of them were designed to systematically characterize PTMs on the endogenous full-length Htt protein. We found that full-length endogenous Htt, which was immunoprecipitated from HD knock-in mouse and human post-mortem brain, is suitable for detection of PTMs by mass spectrometry. Using label-free and mass tag labeling-based approaches, we identified near 40 PTMs, of which half are novel (data are available via ProteomeXchange with identifier PXD005753). Most PTMs were located in clusters within predicted unstructured domains rather than within the predicted α-helical structured HEAT repeats. Using quantitative mass spectrometry, we detected significant differences in the stoichiometry of several PTMs between HD and WT mouse brain. The mass-spectrometry identification and quantitation were verified using phospho-specific antibodies for selected PTMs. To further validate our findings, we introduced individual PTM alterations within full-length Htt and identified several PTMs that can modulate its subcellular localization in striatal cells. These findings will be instrumental in further assembling the Htt PTM framework and highlight several PTMs as potential therapeutic targets for HD.


Subject(s)
Huntingtin Protein/metabolism , Protein Processing, Post-Translational , Animals , Brain/metabolism , Brain Chemistry , Corpus Striatum/pathology , Humans , Huntingtin Protein/chemistry , Huntington Disease/pathology , Mass Spectrometry/methods , Mice , Nerve Tissue Proteins/metabolism , Peptide Hydrolases/chemistry , Phosphorylation , Protein Domains
8.
Elife ; 5: e11184, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-27003594

ABSTRACT

The polyglutamine expansion in huntingtin protein causes Huntington's disease. Here, we investigated structural and biochemical properties of huntingtin and the effect of the polyglutamine expansion using various biophysical experiments including circular dichroism, single-particle electron microscopy and cross-linking mass spectrometry. Huntingtin is likely composed of five distinct domains and adopts a spherical α-helical solenoid where the amino-terminal and carboxyl-terminal regions fold to contain a circumscribed central cavity. Interestingly, we showed that the polyglutamine expansion increases α-helical properties of huntingtin and affects the intramolecular interactions among the domains. Our work delineates the structural characteristics of full-length huntingtin, which are affected by the polyglutamine expansion, and provides an elegant solution to the apparent conundrum of how the extreme amino-terminal polyglutamine tract confers a novel property on huntingtin, causing the disease.


Subject(s)
Huntingtin Protein/chemistry , Huntingtin Protein/metabolism , Peptides/metabolism , Biophysical Phenomena , Circular Dichroism , Mass Spectrometry , Microscopy, Electron , Protein Conformation
9.
Hum Mol Genet ; 24(9): 2442-57, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25574027

ABSTRACT

The CAG repeat expansion in the Huntington's disease gene HTT extends a polyglutamine tract in mutant huntingtin that enhances its ability to facilitate polycomb repressive complex 2 (PRC2). To gain insight into this dominant gain of function, we mapped histone modifications genome-wide across an isogenic panel of mouse embryonic stem cell (ESC) and neuronal progenitor cell (NPC) lines, comparing the effects of Htt null and different size Htt CAG mutations. We found that Htt is required in ESC for the proper deposition of histone H3K27me3 at a subset of 'bivalent' loci but in NPC it is needed at 'bivalent' loci for both the proper maintenance and the appropriate removal of this mark. In contrast, Htt CAG size, though changing histone H3K27me3, is prominently associated with altered histone H3K4me3 at 'active' loci. The sets of ESC and NPC genes with altered histone marks delineated by the lack of huntingtin or the presence of mutant huntingtin, though distinct, are enriched in similar pathways with apoptosis specifically highlighted for the CAG mutation. Thus, the manner by which huntingtin function facilitates PRC2 may afford mutant huntingtin with multiple opportunities to impinge upon the broader machinery that orchestrates developmentally appropriate chromatin status.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion , Alleles , Animals , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Cluster Analysis , Embryonic Stem Cells/metabolism , Gene Deletion , Gene Expression Regulation , Genome-Wide Association Study , Genotype , High-Throughput Nucleotide Sequencing , Histones/metabolism , Huntingtin Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Neural Stem Cells/metabolism , Nuclear Proteins/chemistry , Polycomb Repressive Complex 2/genetics
10.
Obesity (Silver Spring) ; 18(9): 1701-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20094041

ABSTRACT

Although germ-line deletion of c-Jun NH(2)-terminal kinase (JNK) improves overall insulin sensitivity in mice, those studies could not reveal the underlying molecular mechanism and the tissue site(s) in which reduced JNK activity elicits the observed phenotype. Given its importance in nonesterified fatty acids (NEFA) and glucose utilization, we hypothesized that the insulin-sensitive phenotype associated with Jnk deletion originates from loss of JNK function in skeletal muscle. Short hairpin RNA (shRNA)-mediated gene silencing was used to identify the functions of JNK subtypes in regulating energy metabolism and metabolic responses to elevated concentrations of NEFA in C2C12 myotubes, a cellular model of skeletal muscle. We show for the first time that cellular JNK2- and JNK1/JNK2-deficiency divert glucose from oxidation to glycogenesis due to increased glycogen synthase (GS) activity and induction of Pdk4. We further show that JNK2- and JNK1/JNK2-deficiency profoundly increase cellular NEFA oxidation, and their conversion to phospholipids and triglyceride. The increased NEFA utilization was coupled to increased expressions of selective NEFA handling genes including Cd36, Acsl4, and Chka, and enhanced palmitic acid (PA)-dependent suppression of acetyl-CoA carboxylase (Acc). In JNK-intact cells, PA inhibited insulin signaling and glycogenesis. Although silencing Jnk1 and/or Jnk2 prevented PA-induced inhibition of insulin signaling, it did not completely block decreased insulin-mediated glycogenesis, thus indicating JNK-independent pathways in the suppression of glycogenesis by PA. Muscle-specific inhibition of JNK2 (or total JNK) improves the capacity of NEFA utilization and glycogenesis, and is a potential therapeutic target for improving systemic insulin sensitivity in type 2 diabetes (T2D).


Subject(s)
Blood Glucose/metabolism , Fatty Acids, Nonesterified/metabolism , Glycogen/biosynthesis , Insulin Resistance/physiology , JNK Mitogen-Activated Protein Kinases/deficiency , Lipid Metabolism , Muscle Fibers, Skeletal/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Animals , Fatty Acids, Nonesterified/genetics , Gene Silencing , Genes , Glycogen Synthase/metabolism , Insulin Resistance/genetics , Lipid Metabolism/genetics , Lipid Peroxidation , Mice , Oxidation-Reduction , Palmitic Acid/metabolism , Phospholipids/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA, Small Interfering , Sequence Deletion , Signal Transduction , Triglycerides/biosynthesis
11.
Mol Cell Biol ; 28(12): 3932-42, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18391015

ABSTRACT

Mediator is a conserved multisubunit complex that acts as a functional interface between regulatory transcription factors and the general RNA polymerase II initiation apparatus. MED1 is a pivotal component of the complex that binds to nuclear receptors and a broad array of other gene-specific activators. Paradoxically, MED1 is found in only a fraction of the total cellular Mediator complexes, and the mechanisms regulating its binding to the core complex remain unclear. Here, we report that phosphorylation of MED1 by mitogen-activated protein kinase-extracellular signal-regulated kinase (MAPK-ERK) promotes its association with Mediator. We show that MED1 directly binds to the MED7 subunit and that ERK phosphorylation of MED1 enhances this interaction. Interestingly, we found that both thyroid and steroid hormones stimulate MED1 phosphorylation in vivo and that MED1 phosphorylation is required for its nuclear hormone receptor coactivator activity. Finally, we show that MED1 phosphorylation by ERK enhances thyroid hormone receptor-dependent transcription in vitro. Our findings suggest that ERK phosphorylation of MED1 is a regulatory mechanism that promotes MED1 association with Mediator and, as such, may facilitate a novel feed-forward action of nuclear hormones.


Subject(s)
Endodeoxyribonucleases/physiology , Extracellular Signal-Regulated MAP Kinases , Cell Nucleus/metabolism , Endodeoxyribonucleases/chemistry , Gene Expression Regulation , HeLa Cells , Humans , Mediator Complex , Models, Biological , Phosphorylation , Protein Binding , RNA Polymerase II/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Saccharomyces cerevisiae/metabolism , Signal Transduction , Thyroid Hormones/metabolism , Trans-Activators/metabolism
12.
Cancer Res ; 67(9): 4034-41, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483314

ABSTRACT

Androgen receptor (AR) signaling pathways are important for the survival and proliferation of prostate cancer cells. Because AR activity is facilitated by distinct coregulatory factors and complexes, it is conceivable that some of these proteins might also play a role in promoting prostate oncogenesis. The multisubunit Mediator complex is an important coactivator for a broad range of regulatory transcriptional factors including AR, yet its role in prostate cancer is unclear. Here, we used RNA interference to knock down the expression of two integral Mediator components, MED1/TRAP220 and MED17, in prostate cancer cells. MED1/TRAP220 plays a particularly important role in androgen signaling in that it serves as a direct binding target for AR. We found that the knockdown of either subunit markedly decreases transcription from transiently transfected androgen-responsive reporter genes, as well as inhibits androgen-dependent expression of endogenous AR target genes. We show for the first time that loss of either MED1/TRAP220 or MED17 in prostate cancer cells significantly decreases both androgen-dependent and -independent cellular proliferation, inhibits cell cycle progression, and increases apoptosis. Furthermore, we show that MED1/TRAP220 is overexpressed in both AR-positive and -negative prostate cancer cells lines, as well as in 50% (10 of 20) of the clinically localized human prostate cancers we examined, thus suggesting that MED1/TRAP220 hyperactivity may have implications in prostate oncogenesis. In sum, our data suggest that Mediator plays an important coregulatory role in prostate cancer cell proliferation and survival, and therefore, may represent a new target for therapeutic intervention.


Subject(s)
Endodeoxyribonucleases/physiology , Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/pathology , Transcription Factors/physiology , Apoptosis/physiology , Cell Cycle/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Endodeoxyribonucleases/biosynthesis , Endodeoxyribonucleases/genetics , Humans , Male , Mediator Complex Subunit 1 , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA Interference , Transcription Factors/biosynthesis , Transcription Factors/genetics
13.
Biochem Biophys Res Commun ; 325(1): 344-52, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15522239

ABSTRACT

Earlier we have shown that the epidermal growth factor receptor was unable to retain its phospho Tyr signal after the assembly of staphylococcal alpha-hemolysin (alpha-HL). However, the nature of the protein tyrosine phosphatase (PTPase) or its identity is not known. In this report, we demonstrate that the alpha-HL elevates the activity of receptor like protein tyrosine phosphatase sigma (rPTPsigma). The alpha-HL induced dephosphorylation is prominent only in intact A431 cells. The PTPase activity is not inhibited if the alpha-HL treatment precedes PTPase inhibitor treatments. The anti-EGFr immunoprecipitates have exhibited higher PTPase activity after alpha-HL treatment of A431 cells. Interestingly, PTPase activity of anti-EGFr immunoprecipitates from the A431 cells expressing the antisense message of rPTPsigma has not increased despite alpha-HL treatment, confirming the role of rPTPsigma in the dephosphorylation of EGFr. The studies presented here will be useful in understanding the process of signal modulation by the assembly of alpha-HL.


Subject(s)
Bacterial Toxins/metabolism , ErbB Receptors/metabolism , Hemolysin Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Binding Sites , Calcium/metabolism , Cell Line, Tumor , Humans , Immunohistochemistry , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Transforming Growth Factor alpha/metabolism , Vanadates/metabolism
14.
Biochem Biophys Res Commun ; 324(3): 1124-9, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15485671

ABSTRACT

Assembly and penetration of 14-strand beta-barrel of staphylococcal alpha-hemolysin (alpha-HL) is an intriguing phenomenon due to its water soluble property. alpha-HL interacts with the Caveolin-1 of A431 cells for its rapid assembly. A nine amino acid, non-hydrophobic peptide derived from alpha-HL has been shown to block the interaction of alpha-HL with the scaffolding domain of Caveolin-1. alpha-HL's presence was also detected in the Caveolin-1 enriched membrane fractions isolated by ultracentrifugation. Moreover, alpha-HL co-precipitates with Caveolin-1 specifically. In a time-dependent process, alpha-HL associates with the Caveolin-1 and co-localizes with Caveolin-1 that results in an extensive clustering of Caveolin-1 at cell-cell contacts. Mutants of alpha-HL devoid of Caveolin-1 binding motif failed to assemble into heptameric oligomers on the surface of A431 cells. Our data suggest that the conformational changes required to form the heptameric assembly might be triggered at the Caveolin-1 binding motif of alpha-HL.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Caveolins/chemistry , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Amino Acid Motifs , Caveolin 1 , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Kinetics , Membrane Microdomains/chemistry , Methionine/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Rhodamines/chemistry , Time Factors
15.
Biochem Biophys Res Commun ; 324(3): 1130-6, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15485672

ABSTRACT

The assembly of alpha-HL was shown to rapidly progress upon its interaction with Caveolin-1. Treatment of A431 cells with alpha-HL has resulted in clustering of Caveolin-1 at cell-cell contacts. Consistent with this observation, alpha-HL mutants devoid of assembly property have not induced the clustering of Caveolin-1. While cholesterol depletion of A431 cells completely arrests the assembly of alpha-HL, chelation of membrane cholesterol results in its retarded assembly. Interestingly, HT29 cells, with low Caveolin-1 levels, are resistant to alpha-HL attack. Clustering of Caveolin-1, as seen in case of A431 cells, was readily observed in case of HT29 cells transfected with Caveolin-1 construct, thus overexpressing the full length Caveolin-1, upon alpha-HL treatment. A model was constructed to visualize the interactions between alpha-HL and Caveolin-1 which suggests that facile penetration of alpha-HL's beta-barrel might occur through protein-protein interactions with the surrounding 7 alpha-helices of Caveolin-1.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Caveolins/chemistry , Cell Membrane/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Amino Acid Motifs , Caveolin 1 , Caveolins/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Humans , Immunoprecipitation , Kinetics , Methionine/chemistry , Microscopy, Fluorescence , Models, Molecular , Mutation , Nystatin/pharmacology , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Time Factors
16.
Biochem Biophys Res Commun ; 322(1): 29-36, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15313169

ABSTRACT

We have identified a nine amino sequence in alpha-hemolysin (alpha-HL) of Staphylococcus aureus, which binds Caveolin-1. Surface plasmon resonance studies clearly show a concentration dependent interaction of alpha-HL with the scaffolding domain of Caveolin-1. Mutants of alpha-HL, devoid of Caveolin-1 recognition motif, exhibit an alpha-HL like proteinase K digestion profile but the resultant 'half-like' domains are highly susceptible to further proteolysis. They also had the same intrinsic fluorescence emission maxima as the native alpha-HL indicating normal folding. However, these mutants bind 1-anilino-8-naphthalene sulfonic acid probably due to exposure of their hydrophobic core. Moreover, these mutants are non-lytic and do not undergo conformational changes on rabbit RBC membrane surface. Purified Caveolin-1 blocks the hemolysis of RBCs by alpha-HL. Our studies indicate that the Caveolin-1 binding motif of alpha-HL provides stability and shields the hydrophobic core of alpha-HL. The motif also acts as trigger point for initiation of conformational changes.


Subject(s)
Bacterial Toxins/chemistry , Caveolins/chemistry , Hemolysin Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Bacterial Toxins/genetics , Binding Sites , Caveolin 1 , Caveolins/genetics , Dimerization , Hemolysin Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Porosity , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Structure-Activity Relationship
17.
Curr Microbiol ; 44(3): 224-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11821933

ABSTRACT

MutS2 protein of Pyrococcus furiosus has been cloned and over-expressed. Initial characterization reveals that PfuMutS2 possesses a thermostable ATPase activity and a thermostable, nonspecific DNA binding activity. However, PfuMutS2 does not have any detectable mismatch-specific DNA binding activity. It is the first in vitro characterization of an MutS2 family protein.


Subject(s)
Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Pyrococcus furiosus/genetics , Pyrococcus furiosus/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Base Pair Mismatch , Cloning, Molecular , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Stability , Escherichia coli/genetics , Gene Expression , Genes, Archaeal , Molecular Sequence Data , Phenotype , Sequence Homology, Amino Acid , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...