Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Osteoarthritis Cartilage ; 31(5): 613-626, 2023 05.
Article in English | MEDLINE | ID: mdl-36410637

ABSTRACT

OBJECTIVES: Metabolic pathways are a series of chemical reactions by which cells take in nutrient substrates for energy and building blocks needed to maintain critical cellular processes. Details of chondrocyte metabolism and how it rewires during the progression of osteoarthritis (OA) are unknown. This research aims to identify what changes in the energy metabolic state occur in OA cartilage. METHODS: Patient matched OA and non-OA cartilage specimens were harvested from total knee replacement patients. Cartilage was first collected for metabolomics, proteomics, and transcriptomics analyses to study global alterations in OA metabolism. We then determined the metabolic routes by tracking [U-13C] isotope with liquid chromatography-mass spectrometry (LC-MS). We further evaluated cellular bioenergetic profiles by measuring oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) and investigated the effects of low-dose and short-term effects of 2-deoxyglucose (2DG) on chondrocytes. RESULTS: OA chondrocytes showed increased basal ECAR and more lactate production compared to non-OA chondrocytes. [U-13C] glucose labelling revealed that less glucose-derived carbon entered the tricarboxylic acid (TCA) cycle. On the other hand, mitochondrial respiratory rates were markedly decreased in the OA chondrocytes compared to non-OA chondrocytes. These changes were accompanied by decreased cellular ATP production, mitochondrial membrane potential and disrupted mitochondrial morphology. We further demonstrated in vitro that short-term inhibition of glycolysis suppressed matrix degeneration gene expression in chondrocytes and bovine cartilage explants cultured under inflammatory conditions. CONCLUSION: This study represents the first comprehensive comparative analysis of metabolism in OA chondrocytes and lays the groundwork for therapeutic targeting of metabolism in OA.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Animals , Cattle , Chondrocytes/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Energy Metabolism , Glucose/metabolism , Cells, Cultured
2.
Ann Oncol ; 29(5): 1292-1303, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29509840

ABSTRACT

Background: MSR1 repeats are a 36-38 bp minisatellite element that have recently been implicated in the regulation of gene expression, through copy number variation (CNV). Patients and methods: Bioinformatic and experimental methods were used to assess the distribution of MSR1 across the genome, evaluate the regulatory potential of such elements and explore the role of MSR1 elements in cancer, particularly non-familial breast cancer and prostate cancer. Results: MSR1s are predominately located at chromosome 19 and are functionally enriched in regulatory regions of the genome, particularly regions implicated in short-range regulatory activities (H3K27ac, H3K4me1 and H3K4me3). MSR1-regulated genes were found to have specific molecular roles, such as serine-protease activity (P = 4.80 × 10-7) and ion channel activity (P = 2.7 × 10-4). The kallikrein locus was found to contain a large number of MSR1 clusters, and at least six of these showed CNV. An MSR1 cluster was identified within KLK14, with 9 and 11 copies being normal variants. A significant association with the 9-copy allele and non-familial breast cancer was found in two independent populations (P = 0.004; P = 0.03). In the white British population, the minor allele conferred an increased risk of 1.21-3.51 times for all non-familial disease, or 1.7-5.3 times in early-onset disease. The 9-copy allele was also found to be associated with increased risk of prostate cancer in an independent population (odds ratio = 1.27-1.56; P =0.009). Conclusions: MSR1 repeats act as molecular switches that modulate gene expression. It is likely that CNV of MSR1 will affect risk of development of various forms of cancer, including that of breast and prostate. The MSR1 cluster at KLK14 represents the strongest risk factor identified to date in non-familial breast cancer and a significant risk factor for prostate cancer. Analysis of MSR1 genotype will allow development of precise stratification of disease risk and provide a novel target for therapeutic agents.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease , Minisatellite Repeats/genetics , Prostatic Neoplasms/genetics , Age of Onset , Breast Neoplasms/pathology , Case-Control Studies , Computational Biology , DNA Copy Number Variations , Female , Germ-Line Mutation , Histones/genetics , Humans , Kallikreins/genetics , Male , Middle Aged , Multigene Family/genetics , Prostatic Neoplasms/pathology , Risk Assessment/methods
3.
Genes Immun ; 18(3): 184-190, 2017 09.
Article in English | MEDLINE | ID: mdl-28835680

ABSTRACT

Tumor necrosis factor-α (TNF-α) inhibitors are highly effective in suppressing inflammation in ankylosing spondylitis (AS) patients, and operate by suppression of TFN-α and downstream immunological pathways. To determine the mechanisms of action of TNF-α inhibitors in AS patients, we used transcriptomic and bioinformatic approaches on peripheral blood mononuclear cells from AS patients pre and post treatment. We found 656 differentially expressed genes, including the genome-wide significant AS-associated genes, IL6R, NOTCH1, IL10, CXCR2 and TNFRSF1A. A distinctive gene expression profile was found between male and female patients, mainly because of sex chromosome-linked genes and interleukin 17 receptor C, potentially accounting for the differences in clinical manifestation and treatment response between the genders. In addition to immune and inflammation regulatory pathways, like intestinal immune network for IgA production, cytokine-cytokine receptor interaction, Ras signaling pathway, allograft rejection and hematopoietic cell lineage, KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses revealed that infection-associated pathways (influenza A and toxoplasmosis) and metabolism-associated pathways were involved in response to TNF-α inhibitor treatment, providing insight into the mechanism of TNF-α inhibitors.


Subject(s)
Spondylitis, Ankylosing/genetics , Transcriptome , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Female , Gene Expression Profiling , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Middle Aged , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/metabolism
5.
Cell Death Dis ; 4: e562, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23538443

ABSTRACT

Apoptosis induction is an antiviral host response, however, influenza A virus (IAV) infection promotes host cell death. The nucleoprotein (NP) of IAV is known to contribute to viral pathogenesis, but its role in virus-induced host cell death was hitherto unknown. We observed that NP contributes to IAV infection induced cell death and heterologous expression of NP alone can induce apoptosis in human airway epithelial cells. The apoptotic effect of IAV NP was significant when compared with other known proapoptotic proteins of IAV. The cell death induced by IAV NP was executed through the intrinsic apoptosis pathway. We screened host cellular factors for those that may be targeted by NP for inducing apoptosis and identified human antiapoptotic protein Clusterin (CLU) as a novel interacting partner. The interaction between IAV NP and CLU was highly conserved and mediated through ß-chain of the CLU protein. Also CLU was found to interact specifically with IAV NP and not with any other known apoptosis modulatory protein of IAV. CLU prevents induction of the intrinsic apoptosis pathway by binding to Bax and inhibiting its movement into the mitochondria. We found that the expression of IAV NP reduced the association between CLU and Bax in mammalian cells. Further, we observed that CLU overexpression attenuated NP-induced cell death and had a negative effect on IAV replication. Collectively, these findings indicate a new function for IAV NP in inducing host cell death and suggest a role for the host antiapoptotic protein CLU in this process.


Subject(s)
Clusterin/metabolism , Epithelial Cells/metabolism , Influenza A virus/metabolism , RNA-Binding Proteins/metabolism , Respiratory Mucosa/metabolism , Viral Core Proteins/metabolism , Apoptosis , Binding Sites , Cell Line , Clusterin/genetics , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Influenza A virus/genetics , Mitochondria , Nucleocapsid Proteins , Protein Binding , Protein Transport , RNA, Small Interfering/genetics , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Signal Transduction , Viral Core Proteins/antagonists & inhibitors , Viral Core Proteins/genetics , bcl-2-Associated X Protein
6.
Hum Genet ; 129(6): 687-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21465221

ABSTRACT

Genome-wide association studies (GWAS) have identified more than 30 prostate cancer (PrCa) susceptibility loci. One of these (rs2735839) is located close to a plausible candidate susceptibility gene, KLK3, which encodes prostate-specific antigen (PSA). PSA is widely used as a biomarker for PrCa detection and disease monitoring. To refine the association between PrCa and variants in this region, we used genotyping data from a two-stage GWAS using samples from the UK and Australia, and the Cancer Genetic Markers of Susceptibility (CGEMS) study. Genotypes were imputed for 197 and 312 single nucleotide polymorphisms (SNPs) from HapMap2 and the 1000 Genome Project, respectively. The most significant association with PrCa was with a previously unidentified SNP, rs17632542 (combined P = 3.9 × 10(-22)). This association was confirmed by direct genotyping in three stages of the UK/Australian GWAS, involving 10,405 cases and 10,681 controls (combined P = 1.9 × 10(-34)). rs17632542 is also shown to be associated with PSA levels and it is a non-synonymous coding SNP (Ile179Thr) in KLK3. Using molecular dynamic simulation, we showed evidence that this variant has the potential to introduce alterations in the protein or affect RNA splicing. We propose that rs17632542 may directly influence PrCa risk.


Subject(s)
Genetic Predisposition to Disease , Kallikreins/genetics , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Genome-Wide Association Study , Humans , Male , Molecular Dynamics Simulation , Polymorphism, Single Nucleotide , Prostate-Specific Antigen/blood
7.
Clin Exp Allergy ; 35(7): 914-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16008678

ABSTRACT

BACKGROUND: IL-10 is an anti-inflammatory cytokine primarily produced by monocytes and macrophages and plays a key role in asthma. IL10 gene, present in chromosome 1q31-q32, is regulated primarily by transcription and its expression is found to be lower in asthmatics. Earlier studies in diverse populations have identified several promoter polymorphisms. However, no study has been carried out in a genetically untapped large population from the Indian Subcontinent. OBJECTIVE: To investigate the association of the IL10 promoter polymorphisms and asthma in the North Indian population. METHODS: The association study was conducted in a case-control as well as in a family-based design. Polymorphism at -1082 A/G, -819 C/T and -592 C/A nucleotides were genotyped in ethnically matched unrelated patients (N=272), unrelated controls (N=307) and nuclear families (N=164). RESULTS: A suggestive evidence of association was obtained for -1082 A/G polymorphism at the level of alleles and genotypes with asthma in the case-control study (P=0.03). A three-locus haplotype (ATA) was found to be more in asthmatics than in control individuals (P=0.0085). On the other hand, a novel haplotype ATC was found to be more in controls than in asthmatics (P=0.012). These results were further tested in a family-based study. A deviation of transmission was observed for the -1082 A/G polymorphism (P=0.003). The ATA haplotype showed a preferential transmission in asthmatics (P=0.03), while the GCC and a novel ATC haplotype showed preferential non-transmission in asthmatic individuals (P=0.03 and 0.001, respectively). CONCLUSIONS: Using both case-control and family studies, we provide suggestive evidence that the ATA haplotype is positively, whereas GCC and a novel ATC haplotypes of IL10 gene are negatively associated with asthma in Indian population. Our results are interesting enough as to intensify further research to elucidate the functional significance of these single-nucleotide polymorphisms and haplotypes in asthma pathogenesis.


Subject(s)
Asthma/genetics , Interleukin-10/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Adult , Asthma/epidemiology , Case-Control Studies , Family Health , Female , Genotype , Haplotypes/genetics , Humans , Immunoglobulin E/blood , India/epidemiology , Male
9.
Biochemistry ; 40(31): 9115-24, 2001 Aug 07.
Article in English | MEDLINE | ID: mdl-11478878

ABSTRACT

Restrictocin, a member of the fungal ribotoxin family, specifically cleaves a single phosphodiester bond in the 28S rRNA and potently inhibits eukaryotic protein synthesis. Residues Tyr47, His49, Glu95, Phe96, Pro97, Arg120, and His136 have been predicted to form the active site of restrictocin. In this study, we have individually mutated these amino acids to alanine to probe their role in restrictocin structure and function. The role of Tyr47, His49, Arg120, and His136 was further investigated by making additional mutants. Mutating Arg120 or His136 to alanine or the other amino acids rendered the toxin completely inactive, whereas mutating Glu95 to alanine only partially inactivated the toxin. Mutation of Phe96 and Pro97 to Ala had no effect on the activity of restrictocin. The Tyr47 to alanine mutant was inactive in inhibiting protein synthesis, and had a nonspecific ribonuclease activity on 28S rRNA similar to that shown previously for the His49 to Ala mutant. Unlike the His136 to Ala mutant, the double mutants containing Tyr47 or His49 mutated to alanine along with His136 did not compete with restrictocin to cause a significant reduction in the extent of cleavage of 28S rRNA. In a model of restrictocin and a 29-mer RNA substrate complex, residues Tyr47, His49, Glu95, Arg120, and His136 were found to be near the cleavage site on RNA. It is proposed that in restrictocin Glu95 and His136 are directly involved in catalysis, Arg120 is involved in the stabilization of the enzyme-substrate complex, Tyr47 provides structural stability to the active site, and His49 determines the substrate specificity.


Subject(s)
Allergens , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Mycotoxins/chemistry , Mycotoxins/metabolism , RNA, Ribosomal, 28S/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism , Animals , Antigens, Plant , Arginine/chemistry , Arginine/genetics , Circular Dichroism , Escherichia coli/genetics , Fungal Proteins/genetics , Fungal Proteins/pharmacology , Glutamic Acid/chemistry , Glutamic Acid/genetics , HeLa Cells , Histidine/chemistry , Histidine/genetics , Humans , Hydrolysis , Models, Molecular , Mutagenesis, Site-Directed , Mycotoxins/genetics , Mycotoxins/pharmacology , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Reticulocytes/drug effects , Reticulocytes/metabolism , Ribonucleases/genetics , Ribonucleases/pharmacology , Substrate Specificity/genetics , Tyrosine/chemistry , Tyrosine/genetics
10.
J Biol Chem ; 276(27): 24978-84, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11342552

ABSTRACT

Mammalian ribonucleases interact very strongly with the intracellular ribonuclease inhibitor (RI). Eukaryotic cells exposed to mammalian ribonucleases are protected from their cytotoxic action by the intracellular inhibition of ribonucleases by RI. Human pancreatic ribonuclease (HPR) is structurally and functionally very similar to bovine RNase A and interacts with human RI with a high affinity. In the current study, we have investigated the involvement of Lys-7, Gln-11, Asn-71, Asn-88, Gly-89, Ser-90, and Glu-111 in HPR in its interaction with human ribonuclease inhibitor. These contact residues were mutated either individually or in combination to generate mutants K7A, Q11A, N71A, E111A, N88R, G89R, S90R, K7A/E111A, Q11A/E111A, N71A/E111A, K7A/N71A/E111A, Q11A/N71A/E111A, and K7A/Q11A/N71A/E111A. Out of these, eight mutants, K7A, Q11A, N71A, S90R, E111A, Q11A/E111A, N71A/E111A, and K7A/N71A/E111A, showed an ability to evade RI more than the wild type HPR, with the triple mutant K7A/N71A/E111A having the maximum RI resistance. As a result, these variants exhibited higher cytotoxic activity than wild type HPR. The mutation of Gly-89 in HPR produced no change in the sensitivity of HPR for RI, whereas it has been reported that mutating the equivalent residue Gly-88 in RNase A yielded a variant with increased RI resistance and cytotoxicity. Hence, despite its considerable homology with RNase A, HPR shows differences in its interaction with RI. We demonstrate that interaction between human pancreatic ribonuclease and RI can be disrupted by mutating residues that are involved in HPR-RI binding. The inhibitor-resistant cytotoxic HPR mutants should be useful in developing therapeutic molecules.


Subject(s)
Enzyme Inhibitors/metabolism , Placental Hormones/metabolism , Ribonuclease, Pancreatic/metabolism , Amino Acid Substitution , Animals , Blotting, Western , Catalysis , Cattle , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Humans , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Conformation , Ribonuclease, Pancreatic/genetics
11.
Eur J Biochem ; 267(6): 1777-83, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712610

ABSTRACT

Restrictocin, produced by the fungus Aspergillus restrictus, is a highly specific ribonucleolytic toxin which cleaves a single phosphodiester bond between G4325 and A4326 in the 28S rRNA. It is a nonglycosylated, single-chain, basic protein of 149 amino acids. The putative catalytic site of restrictocin includes Tyr47, His49, Glu95, Arg120 and His136. To map the catalytic activity in the restrictocin molecule, and to study the role of N- and C-terminus in its activity, we have systematically deleted amino-acid residues from both the termini. Three N-terminal deletions removing 8, 15 and 30 amino acids, and three C-terminal deletions lacking 4, 6, and 11 amino acids were constructed. The deletion mutants were expressed in Escherichia coli, purified to homogeneity and functionally characterized. Removal of eight N-terminal or four C-terminal amino acids rendered restrictocin partially inactive, whereas any further deletions from either end resulted in the complete inactivation of the toxin. The study demonstrates that intact N- and C-termini are required for the optimum functional activity of restrictocin.


Subject(s)
Allergens , Aspergillus/metabolism , Fungal Proteins/metabolism , Mycotoxins/metabolism , Ribonucleases/metabolism , Amino Acids/chemistry , Antigens, Plant , Catalysis , Circular Dichroism , Escherichia coli , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/pharmacology , HeLa Cells , Humans , Mutagenesis, Site-Directed , Mycotoxins/chemistry , Mycotoxins/genetics , Mycotoxins/pharmacology , Protein Biosynthesis/drug effects , Protein Engineering , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/pharmacology , Ribosomes/drug effects , Sequence Deletion , Structure-Activity Relationship
12.
Biochem J ; 345 Pt 2: 247-54, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10620501

ABSTRACT

Chimaeric toxins have considerable therapeutic potential to treat various malignancies. We have previously used the fungal ribonucleolytic toxin restrictocin to make chimaeric toxins in which the ligand was fused at either the N-terminus or the C-terminus of the toxin. Chimaeric toxins containing ligand at the C-terminus of restrictocin were shown to be more active than those having ligand at the N-terminus of the toxin. Here we describe the further engineering of restrictocin-based chimaeric toxins, anti-TFR(scFv)-restrictocin and restrictocin-anti-TFR(scFv), containing restrictocin and a single chain fragment variable (scFv) of a monoclonal antibody directed at the human transferrin receptor (TFR), to enhance their cell-killing activity. To promote the independent folding of the two proteins in the chimaeric toxin, a linear flexible peptide, Gly-Gly-Gly-Gly-Ser, was inserted between the toxin and the ligand to generate restrictocin-linker-anti-TFR(scFv) and anti-TFR(scFv)-linker-restrictocin. A 12-residue spacer, Thr-Arg-His-Arg-Gln-Pro-Arg-Gly-Trp-Glu-Gln-Leu, containing the recognition site for the protease furin, was incorporated between the toxin and the ligand to generate restrictocin-spacer-anti-TFR(scFv) and anti-TFR(scFv)-spacer-restrictocin. The incorporation of the proteolytically cleavable spacer enhanced the cell-killing activity of both constructs by 2-30-fold depending on the target cell line. However, the introduction of linker improved the cytotoxic activity only for anti-TFR(scFv)-linker-restrictocin. The proteolytically cleavable spacer-containing chimaeric toxins had similar cytotoxic activities irrespective of the location of the ligand on the toxin and they were found to release the restrictocin fragment efficiently on proteolysis in vitro.


Subject(s)
Allergens , Antibiotics, Antineoplastic/pharmacology , Fungal Proteins/pharmacology , Immunoglobulin Fragments/pharmacology , Immunotoxins/pharmacology , Receptors, Transferrin/immunology , Ribonucleases/pharmacology , Subtilisins/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Antigens, Plant , Cell Line , Circular Dichroism , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Furin , Humans , Immunoglobulin Fragments/genetics , Immunotoxins/chemistry , Immunotoxins/genetics , Immunotoxins/metabolism , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Binding , Protein Engineering , Protein Structure, Secondary , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/metabolism , Toxicity Tests , Trypsin/pharmacology
13.
Biochemistry ; 38(31): 10052-8, 1999 Aug 03.
Article in English | MEDLINE | ID: mdl-10433712

ABSTRACT

Restrictocin, produced by the fungus Aspergillus restrictus, belongs to the group of ribonucleolytic toxins called ribotoxins. It specifically cleaves a single phosphodiester bond in a conserved stem and loop structure in the 28S rRNA of large ribosomal subunit and potently inhibits eukaryotic protein synthesis. Restrictocin contains 149 amino acid residues and includes four cysteines at positions 5, 75, 131, and 147. These cysteine residues are involved in the formation of two disulfide bonds, one between Cys 5 and Cys 147 and another between Cys 75 and Cys 131. In the current study, all four cysteine residues were changed to alanine individually and in different combinations by site-directed mutagenesis so as to remove one or both the disulfides. The mutants were expressed and purified from Escherichia coli. Removal of any cysteine or any one of the disulfide bonds individually did not affect the ability of the toxin to specifically cleave the 28S rRNA or to inhibit protein synthesis in vitro. However, the toxin without both disulfide bonds completely lost both ribonucleolytic and protein synthesis inhibition activities. The active mutants, containing only one disulfide bond, exhibited relatively high susceptibility to trypsin digestion. Thus, none of the four cysteine residues is directly involved in restrictocin catalysis; however, the presence of any one of the two disulfide bonds is absolutely essential and sufficient to maintain the enzymatically active conformation of restrictocin. For maintenance of the unique stability displayed by the native toxin, both disulfide bonds are required.


Subject(s)
Allergens , Aspergillus/enzymology , Cysteine/chemistry , Cytotoxins/chemistry , Disulfides/chemistry , Fungal Proteins/chemistry , Ribonucleases/chemistry , Amino Acid Substitution/genetics , Animals , Antigens, Plant , Aspergillus/genetics , Cell-Free System/drug effects , Cysteine/genetics , Cytotoxins/genetics , Cytotoxins/metabolism , Cytotoxins/toxicity , Enzyme Stability/genetics , Escherichia coli/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungal Proteins/toxicity , Hot Temperature , Hydrolysis , Mutagenesis, Site-Directed , Protein Synthesis Inhibitors/pharmacology , Rabbits , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Ribonucleases/genetics , Ribonucleases/metabolism , Ribonucleases/toxicity , Structure-Activity Relationship , Trypsin/metabolism
15.
Biochemistry ; 36(44): 13693-9, 1997 Nov 04.
Article in English | MEDLINE | ID: mdl-9354640

ABSTRACT

Restrictocin is a small basic protein produced by the fungus Aspergillus restrictus. It potently inhibits protein synthesis in eukaryotic cells by specifically cleaving a single phosphodiester bond in 28S rRNA. A histidine residue at position 49 in restrictocin has been implicated in its active site. A mutant of restrictocin in which the histidine at position 49 was changed to an alanine was constructed by site-directed mutagenesis, and the protein was expressed in Escherichia coli. The mutant and the wild type proteins were found to be structurally identical. Unlike restrictocin, the restrictocin H49A mutant did not cleave the ribosomal RNA specifically at the target phosphodiester bond; instead, it extensively degraded the RNA substrate with altered specificity. The mutant exhibited a high ribonuclease activity compared to restrictocin on yeast tRNA, and poly(U) and poly(C). The mutant also poorly inhibited protein synthesis in eukaryotic cells as well as in a cell free system. We therefore propose that histidine 49 of restrictocin is not involved per se in the enzymatic activity; however, it does play a crucial role in the specific recognition of the target sequence by restrictocin.


Subject(s)
Allergens , Amino Acid Substitution/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mycotoxins/genetics , Mycotoxins/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism , Alanine/genetics , Animals , Antigens, Plant , Fungal Proteins/isolation & purification , Genetic Vectors , HeLa Cells , Histidine/genetics , Humans , Kinetics , Mutagenesis, Site-Directed , Mycotoxins/isolation & purification , Protein Synthesis Inhibitors/metabolism , Protein Synthesis Inhibitors/toxicity , RNA, Fungal/metabolism , RNA, Ribosomal, 28S/metabolism , Rabbits , Ribonucleases/isolation & purification , Substrate Specificity/drug effects , Substrate Specificity/genetics
16.
Biochem J ; 324 ( Pt 3): 815-22, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9210405

ABSTRACT

Restrictocin is a ribonucleolytic toxin produced by the fungus Aspergillus restrictus. Two chimaeric toxins containing restrictocin directed at the human transferrin receptor have been constructed. Anti-TFR(scFv)-restrictocin is encoded by a gene produced by fusing the DNA encoding a single-chain antigen-combining region (scFv) of a monoclonal antibody, directed at the human transferrin receptor, at the 5' end of that encoding restrictocin. The other chimaeric toxin, restrictocin-anti-TFR(scFv), is encoded by a gene fusion containing the DNA encoding the single-chain antigen-combining region of antibody to human transferrin receptor at the 3' end of the DNA encoding restrictocin. These gene fusions were expressed in Escherichia coli, and fusion proteins purified from the inclusion bodies by simple chromatography techniques to near-homogeneity. The two chimaeric toxins were found to be equally active in inhibiting protein synthesis in a cell-free in vitro translation assay system. The chimaeric toxins were selectively toxic to the target cells in culture with potent cytotoxic activities. However, restrictocin-anti-TFR(scFv) was more active than anti-TFR(scFv)-restrictocin on all cell lines studied. By using protease and metabolic inhibitors, it can be shown that, to manifest their cytotoxic activity, the restrictocin-containing chimaeric toxins need to be proteolytically processed intracellularly and the free toxin or a fragment thereof thus generated is translocated to the target via a route involving the Golgi apparatus.


Subject(s)
Allergens , Cytotoxins/genetics , Fungal Proteins/genetics , Recombinant Fusion Proteins/genetics , Ribonucleases/genetics , Antigens, Plant , Cloning, Molecular , Cytotoxins/metabolism , Cytotoxins/toxicity , Drug Interactions , Fungal Proteins/metabolism , Fungal Proteins/toxicity , Humans , Kinetics , Protease Inhibitors/pharmacology , Protein Binding , Protein Synthesis Inhibitors/toxicity , Receptors, Transferrin/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/toxicity , Ribonucleases/metabolism , Ribonucleases/toxicity , Tumor Cells, Cultured
17.
FEBS Lett ; 407(3): 275-9, 1997 May 05.
Article in English | MEDLINE | ID: mdl-9175867

ABSTRACT

Targeted toxins represent a new approach to specific cytocidal therapy. The ribonucleolytic protein toxin restrictocin is a potent protein synthesis inhibitor produced by the fungus Aspergillus restrictus. In the present study we have constructed two restrictocin based chimeric toxins where human transforming growth factor alpha (TGF alpha) has been used as a ligand. TGF alpha is a single chain polypeptide, which binds to epidermal growth factor receptor (EGFR) and causes proliferation in a large number of cancers. The ligand has been separately fused either at the amino terminus or carboxyl terminus of restrictocin, giving rise to TGF alpha-restrictocin and restrictocin-TGF alpha respectively. The fusion proteins were overexpressed in Escherichia coli and purified from inclusion bodies by a denaturation-renaturation protocol. Both the chimeric toxins actively inhibited eukaryotic protein synthesis in a cell free in vitro translation assay system. These chimeric toxins selectively killed human epidermal growth factor receptor positive target cells in culture. Among the two proteins, restrictocin-TGF alpha was more active than TGF alpha-restrictocin on all the cell lines studied.


Subject(s)
Allergens , Cytotoxins/toxicity , ErbB Receptors/drug effects , Fungal Proteins/toxicity , Mycotoxins/toxicity , Ribonucleases/toxicity , Animals , Antigens, Plant , Cell Line , Cytotoxins/chemistry , Cytotoxins/genetics , ErbB Receptors/metabolism , Escherichia coli/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Humans , In Vitro Techniques , Mice , Mycotoxins/chemistry , Mycotoxins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/toxicity , Ribonucleases/chemistry , Ribonucleases/genetics , Transforming Growth Factor alpha/metabolism
18.
Gene ; 190(1): 31-5, 1997 Apr 29.
Article in English | MEDLINE | ID: mdl-9185846

ABSTRACT

alpha-Sarcin is a ribonucleolytic protein secreted by the mold Aspergillus giganteus. DNA encoding alpha-sarcin was isolated from the host and cloned into T7 promoter based E. coli expression vectors. Using bacterial outer membrane protein A (OmpA) signal sequence, properly processed recombinant (re-) protein was secreted into the culture medium while in the absence of a signal sequence protein remained insoluble in the bacterial inclusion bodies. The re-alpha-sarcin was purified to homogeneity by simple chromatographic techniques both from the insoluble and soluble sources with respective yields of 40-50 microg/ml and 2-3 microg/ml. The re-ribotoxin was functionally as active as the native toxin and preserved its specificity. The re-alpha-sarcin was used in the construction of an active immunotoxin targeted at the human cancer cells overexpressing transferrin receptor (TFR).


Subject(s)
Aspergillus/genetics , Endoribonucleases/genetics , Fungal Proteins , Immunotoxins/genetics , Cloning, Molecular , Escherichia coli/genetics
19.
Eur J Biochem ; 245(2): 465-9, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9151980

ABSTRACT

Mammalian ribonucleases constitute one of the fastest evolving protein families in nature. The addition of a four-residue carboxyl-terminal tail: Glu-Asp-Ser-Thr (EDST) in human pancreatic ribonuclease (HPR) in comparison with bovine pancreatic RNase (RNase A) could have adaptive significance in humans. We have cloned and expressed human pancreatic ribonuclease in Escherichia coli to probe the influence of the four-residue extension and neighboring C-terminal residues on the biochemical properties of the enzyme. Removal of the C-terminal extension from HPR yielded an enzyme, HPR-(1-124)-peptide, with enhanced ability to cleave poly(C). HPR-(1-124)-peptide also exhibited a steep increase in thermal stability mimicking that known for RNase A. Wild-type HPR had significantly low thermal stability compared to RNase A. The study identifies the C-terminal boundary in the human pancreatic ribonuclease required for efficient catalysis.


Subject(s)
Ribonuclease, Pancreatic/chemistry , Amino Acid Sequence , Animals , Cattle , Circular Dichroism , Cloning, Molecular , Enzyme Stability , Escherichia coli , Hot Temperature , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Structure-Activity Relationship
20.
FEBS Lett ; 392(3): 259-62, 1996 Sep 02.
Article in English | MEDLINE | ID: mdl-8774857

ABSTRACT

Restrictocin is a toxin produced by the fungus Aspergillus restritus. The DNA coding for restrictocin was isolated from the host by polymerase chain reaction and cloned into a T7 promoter-based expression vector. The protein was overproduced in Escherichia coli and remained insoluble in the cell in the form of inclusion bodies. Recombinant restrictocin was purified in large amounts, by a simple denaturation-renaturation protocol involving a redox system, with typical yields of 45 mg/l of original culture. Restrictocin could be secreted into the bacterial medium using ompA, pelB and LTB signal sequences. Among the three signal sequences, ompA was found to be the most efficient in secreting the recombinant protein. The protein secreted into the extracellular medium was properly processed as evident by the amino-terminal sequencing. Recombinant restrictocin was readily purified to homogeneity from either the medium or inclusion bodies by simple chromatographic techniques and was found to be functionally as active as the native fungal protein in inhibiting the eukaryotic translation.


Subject(s)
Allergens , Escherichia coli/metabolism , Fungal Proteins/metabolism , Ribonucleases/metabolism , Amino Acid Sequence , Animals , Antigens, Plant , Circular Dichroism , Escherichia coli/genetics , Fungal Proteins/isolation & purification , Fungal Proteins/pharmacology , Globins/antagonists & inhibitors , Molecular Sequence Data , Plasmids/chemistry , Plasmids/genetics , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reticulocytes/drug effects , Ribonucleases/isolation & purification , Ribonucleases/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...