Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Int J Mol Sci ; 24(13)2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37445686

ABSTRACT

Bacterial superantigens (SAgs) are effective T-cell stimulatory molecules that lead to massive cytokine production. Superantigens crosslink between MHC class II molecules on the Antigen Presenting Cells (APC) and TCR on T-cells. This enables them to activate up to 20% of resting T cells, whilst conventional antigen presentation results in the activation of 0.001-0.0001% of the T cell population. These biological properties of superantigens make them attractive for use in immunotherapy. Previous studies have established the effectiveness of superantigens as therapeutic agents. This, however, was achieved with severe side effects due to the high lethality of the native toxins. Our study aims to produce superantigen-based peptides with minimum or no lethality for safer cancer treatment. In previous work, we designed and synthesized twenty overlapping SPEA-based peptides and successfully mapped regions in SPEA superantigen, causing a vasodilatory response. We screened 20 overlapping SPEA-based peptides designed and synthesized to cover the whole SPEA molecule for T-cell activation and tumor-killing ability. In addition, we designed and synthesized tumor-targeted superantigen-based peptides by fusion of TGFαL3 either from the N' or C' terminal of selected SPEA-based peptides with an eight-amino acid flexible linker in between. Our study identified parts of SPEA capable of stimulating human T-cells and producing different cytokines. We also demonstrated that the SPEA-based peptide conjugate binds specifically to cancer cells and can kill this cancer. Peptides induce T-cell activation, and tumor killing might pave the way for safer tumor-targeted superantigens (TTS). We proposed the combination of our new superantigen-based peptide conjugates with other immunotherapy techniques for effective and safer cancer treatment.


Subject(s)
Neoplasms , Superantigens , Humans , Peptides/pharmacology , T-Lymphocytes , Neoplasms/therapy , Immunotherapy , Enterotoxins
3.
Technol Cancer Res Treat ; 20: 15330338211057371, 2021.
Article in English | MEDLINE | ID: mdl-34802309

ABSTRACT

Objectives: Aminopeptidase N (APN) is an enzyme highly expressed in metastatic cancers and could be used in targeted cancer therapy. Our previous work showed the successful construction of CNGRC-carboxypeptidase G2 (CPG2) and CNGRC-CPG2-CNGRC fusion proteins. Our conjugates and prodrugs were effective in targeting high APN-expressing cancer cells. In the present study, we aim to produce long-acting fusion proteins to overcome 2 of the main drawbacks of antibody-directed enzyme prodrug therapy. Methods: N-terminal and N-, C-terminal fusion CPG2, CNGRC-CPG2, and CNGRC-CPG2-CNGRC, respectively, were PEGylated using polyethylene glycol (PEG) maleimide (40K). We examined the effect of PEGylation on the therapeutic efficacy of the new products. The resulting PEGylated fusion proteins were tested for their stability, ex vivo immunotoxicity, binding capacity to their target on high HT1080, and low A549 APN-expressing cells. The catalytic activity of the resulting PEGylated fusion CPG2 proteins was investigated. Pro-drug "ZD2767P" cytotoxic effect in association with PEG CPG2-CNGRC fusion proteins on cancer cells was studied. Results: Our work demonstrated that the properties of the PEGylated single-fused proteins were significantly improved over that of un-PEGylated fused CPG2, and its kinetic activity and APN-binding affinity were not negatively affected by the PEGylation. Significantly, The PEGylated single-fused CPG2 had lower immunogenicity than the un-PEGylated CPG2. Our results, however, were different in the case of the PEGylated double-fused CPG2. Although its stability in human serum under physiological conditions was not significantly affected, the kinetic activity and its binding affinity to their cellular marker (APN) were substantially reduced. When the study was performed with high and low APN-expressing cancer cell lines, using the prodrug ZD2767p, the PEGylated fusion CPG2 demonstrated cancer cell killing effects. Conclusion: We have successfully produced PEGylated-CNGRC-CPG2, which is bioactive and with lower immunogenicity in ligand-directed enzyme prodrug therapy for cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides, Cyclic , Prodrugs/pharmacology , Recombinant Fusion Proteins/pharmacology , gamma-Glutamyl Hydrolase , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Ligands , Molecular Targeted Therapy , Peptides, Cyclic/chemistry , Polyethylene Glycols , Prodrugs/chemistry , Recombinant Fusion Proteins/chemistry , Spectrum Analysis , gamma-Glutamyl Hydrolase/chemistry
4.
Biomed Pharmacother ; 144: 112260, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607105

ABSTRACT

Abnormal structural and molecular changes in malignant tissues were thoroughly investigated and utilized to target tumor cells, hence rescuing normal healthy tissues and lowering the unwanted side effects as non-specific cytotoxicity. Various ligands for cancer cell specific markers have been uncovered and inspected for directional delivery of the anti-cancer drug to the tumor site, in addition to diagnostic applications. Over the past few decades research related to the ligand targeted therapy (LTT) increased tremendously aiming to treat various pathologies, mainly cancers with well exclusive markers. Malignant tumors are known to induce elevated levels of a variety of proteins and peptides known as cancer "markers" as certain antigens (e.g., Prostate specific membrane antigen "PSMA", carcinoembryonic antigen "CEA"), receptors (folate receptor, somatostatin receptor), integrins (Integrin αvß3) and cluster of differentiation molecules (CD13). The choice of an appropriate marker to be targeted and the design of effective ligand-drug conjugate all has to be carefully selected to generate the required therapeutic effect. Moreover, since some tumors express aberrantly high levels of more than one marker, some approaches investigated targeting cancer cells with more than one ligand (dual or multi targeting). We aim in this review to report an update on the cancer-specific receptors and the vehicles to deliver cytotoxic drugs, including recent advancements on nano delivery systems and their implementation in targeted cancer therapy. We will discuss the advantages and limitations facing this approach and possible solutions to mitigate these obstacles. To achieve the said aim a literature search in electronic data bases (PubMed and others) using keywords "Cancer specific receptors, cancer specific antibody, tumor specific peptide carriers, cancer overexpressed proteins, gold nanotechnology and gold nanoparticles in cancer treatment" was carried out.


Subject(s)
Antineoplastic Agents/administration & dosage , Cancer Vaccines/therapeutic use , Drug Carriers , Drug Resistance, Neoplasm , Genetic Therapy , Neoplasms/therapy , Precision Medicine , Animals , Antineoplastic Agents/metabolism , CRISPR-Cas Systems , Cancer Vaccines/adverse effects , Drug Compounding , Drug Resistance, Neoplasm/genetics , Humans , Molecular Targeted Therapy , Nanoparticles , Nanotechnology , Neoplasms/diagnosis , Neoplasms/genetics , Neoplasms/immunology
5.
Org Lett ; 22(14): 5396-5400, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32584589

ABSTRACT

A concise and modular total synthesis of the highly potent N14-desacetoxytubulysin H (1) has been accomplished in 18 steps in an overall yield of up to 30%. Our work highlights the complexity-augmenting and route-shortening power of diastereoselective multicomponent reaction (MCR) as well as the role of bulky ligands to perfectly control both the regioselective and diastereoselective synthesis of tubuphenylalanine in just two steps. The total synthesis not only provides an operationally simple and step economy but will also stimulate major advances in the development of new tubulysin analogues.

6.
Oncotarget ; 11(6): 619-633, 2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32110281

ABSTRACT

The sequence asparagine-glycine arginine (NGR), flanked by Cysteine (Cys) residues so as to form a disulfide-bridge (CNGRC), has previously been found to target and bind specifically to aminopeptidase N (APN), which is highly expressed on the surface of tumor cells. The goal of this study was to develop and evaluate the potential of fusion proteins carrying the CNGRC sequence linked to the enzyme carboxypeptidase G2 (CPG2) for targeted cancer therapy. We refer to this strategy as ligand-directed enzyme prodrug therapy (LDEPT). We constructed two forms of the CNGRC-CPG2 fusions, containing one or two copies of the cyclic NGR motif and designated CNGRC-CPG2 (X-CPG2) and CNGRC-CPG2-CNGRC (X-CPG2-X), respectively. In vitro binding assays of the purified constructs showed that both X-CPG2 and X-CPG2-X bound with high affinity to cancer cells expressing high levels of APN, compared to their binding to cells expressing low levels of APN. Further in vitro studies of the constructs to assess the therapeutic potential of LDEPT were carried out using cells expressing high and low levels of APN. Using methotrexate, it was demonstrated that cancer cell survival was significantly higher in the presence of the fusion proteins, due to the hydrolysis of this cytotoxic drug by CPG2. Conversely, when the prodrug ZD2767P was used, cancer cell killing was higher in the presence of the fused CPG2 constructs than in their absence, which is consistent with CPG2-mediated release of the cytotoxic drug from the prodrug. Furthermore, the doubly-fused CPG2 construct (X-CPG2-X) was significantly more effective than the singly-fused construct (X-CPG2).

7.
Biomed Pharmacother ; 125: 110009, 2020 May.
Article in English | MEDLINE | ID: mdl-32106381

ABSTRACT

BACKGROUND: Until recently, patients who have the same type and stage of cancer all receive the same treatment. It has been established, however, that individuals with the same disease respond differently to the same therapy. Further, each tumor undergoes genetic changes that cause cancer to grow and metastasize. The changes that occur in one person's cancer may not occur in others with the same cancer type. These differences also lead to different responses to treatment. Precision medicine, also known as personalized medicine, is a strategy that allows the selection of a treatment based on the patient's genetic makeup. In the case of cancer, the treatment is tailored to take into account the genetic changes that may occur in an individual's tumor. Precision medicine, therefore, could be defined in terms of the targets involved in targeted therapy. METHODS: A literature search in electronic data bases using keywords "cancer targeted therapy, personalized medicine and cancer combination therapies" was conducted to include papers from 2010 to June 2019. RESULTS: Recent developments in strategies of targeted cancer therapy were reported. Specifically, on the two types of targeted therapy; first, immune-based therapy such as the use of immune checkpoint inhibitors (ICIs), immune cytokines, tumor-targeted superantigens (TTS) and ligand targeted therapeutics (LTTs). The second strategy deals with enzyme/small molecules-based therapies, such as the use of a proteolysis targeting chimera (PROTAC), antibody-drug conjugates (ADC) and antibody-directed enzyme prodrug therapy (ADEPT). The precise targeting of the drug to the gene or protein under attack was also investigated, in other words, how precision medicine can be used to tailor treatments. CONCLUSION: The conventional therapeutic paradigm for cancer and other diseases has focused on a single type of intervention for all patients. However, a large literature in oncology supports the therapeutic benefits of a precision medicine approach to therapy as well as combination therapies.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Precision Medicine , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Clinical Studies as Topic , Combined Modality Therapy , Disease Management , Disease Susceptibility , Drug Evaluation, Preclinical , Host-Pathogen Interactions , Humans , Neoplasms/etiology , Neoplasms/mortality , Neoplasms/pathology , Prognosis , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
8.
Biomed Pharmacother ; 115: 108905, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31060004

ABSTRACT

Superantigens (SAgs) are a class of antigens that cause non-specific activation of T-cells resulting in polyclonal T cell activation and massive cytokine release and causing symptoms similar to sepsis, e.g. hypotension and subsequent hyporeactivity. We investigated the direct effect of SAgs on vascular tone using two recombinant SAgs, SEA and SPEA. The roles of Nitric Oxide (NO) and potentially hyperpolarization, which is dependent on the K+ channel activation, were also explored. The data show that SEA and SPEA have direct vasodilatory effects that were in part NO-dependent, but completely dependent on activation of K+ channels. Our work also identified the functional regions of one of the superantigens, SPEA, that are involved in causing the vasodilation and possible hypotension. A series of 20 overlapping peptides, spanning the entire sequence of SPEA, were designed and synthesized. The vascular response of each peptide was measured, and the active peptides were identified. Our results implicate the regions, (61-100), (101-140) and (181-220) which cause the vasodilation and possible hypotension effects of SPEA. The data also shows that the peptide 181-220 exert the highest vasodilation effect. This work therefore, demonstrates the direct effect of SAgs on vascular tone and identify the active region causing this vasodilation. We propose that these three peptides could be effective novel antihypertensive drugs. We also overexpressed, in E.coli, four superantigens from codon optimized genes.


Subject(s)
Lymphocyte Activation/drug effects , Neoplasms/drug therapy , Peptides/pharmacology , Superantigens/pharmacology , T-Lymphocytes/drug effects , Vasodilation/drug effects , Animals , Escherichia coli/genetics , Immunotherapy , Muscle, Smooth, Vascular/drug effects , Neoplasms/immunology , Peptides/genetics , Sheep , Superantigens/genetics , T-Lymphocytes/immunology
9.
Biomed Pharmacother ; 112: 108725, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30970523

ABSTRACT

Glucarpidase, also known as carboxypeptidase G2, is a Food and Drug Administration-approved enzyme used in targeted cancer strategies such as antibody-directed enzyme prodrug therapy (ADEPT). It is also used in drug detoxification when cancer patients have excessive levels of the anti-cancer agent methotrexate. The application of glucarpidase is limited by its potential immunogenicity and limited catalytic efficiency. To overcome these pitfalls, mutagenesis was applied to the glucarpidase gene of Pseudomonas sp. strain RS-16 to isolate three novels "biobetter" variants with higher specific enzyme activity. DNA sequence analysis of the genes for the variants showed that each had a single point mutation, resulting in the amino acid substitutions: I100 T, G123S and T239 A. Km, Vmax and Kcat measurements confirmed that each variant had increased catalytic efficiency relative to wild type glucarpidase. Additionally, circular dichroism studies indicated that they had a higher alpha-helical content relative to the wild type enzyme. However, three different software packages predicted that they had reduced protein stability, which is consistent with having higher activities as a tradeoff. The novel glucarpidase variants presented in this work could pave the way for more efficient drug detoxification and might allow dose escalation during chemotherapy. They also have the potential to increase the efficiency of ADEPT and to reduce the number of treatment cycles, thereby reducing the risk that patients will develop antibodies to glucarpidase.


Subject(s)
Drug Design , Prodrugs , Pseudomonas putida/genetics , gamma-Glutamyl Hydrolase/genetics , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/pharmacokinetics , Cloning, Molecular , Enzyme Stability , Enzyme Therapy/methods , Methotrexate/adverse effects , Methotrexate/pharmacokinetics , Models, Molecular , Neoplasms/drug therapy , Neoplasms/immunology , Point Mutation , Prodrugs/administration & dosage , Prodrugs/therapeutic use , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , gamma-Glutamyl Hydrolase/immunology , gamma-Glutamyl Hydrolase/therapeutic use
10.
Biomed Pharmacother ; 113: 108750, 2019 May.
Article in English | MEDLINE | ID: mdl-30849643

ABSTRACT

Protein therapeutics play a significant role in treating many diseases. They, however, suffer from patient's proteases degradation and antibody neutralization which lead to short plasma half-lives. One of the ways to overcome these pitfalls is the frequent injection of the drug albeit at the cost of patient compliance which affects the quality of life of patients. There are several techniques available to extend the half-life of therapeutics. Two of the most common protocols are PEGylation and fusion with human serum albumin. These two techniques improve stability, reduce immunogenicity, and increase drug resistance to proteases. These factors lead to the reduction of injection frequency which increases patient compliance and improve quality of life. Both techniques have already been used in many FDA approved drugs. This review describes many technologies to produce long-acting drugs with the attention of PEGylation and the genetic fusion with human serum albumin. The report also discusses the latest modified therapeutics in the field and their application in cancer therapy. We compare the modification methods and discuss the pitfalls of these modified drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Delayed-Action Preparations , Half-Life , Humans , Polyethylene Glycols/chemistry , Quality of Life , Serum Albumin, Human/chemistry
11.
Eur J Pharm Sci ; 127: 79-91, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30343151

ABSTRACT

Recombinant glucarpidase (formerly: Carboxypeptidase G2, CPG2) is used in Antibody Directed Enzyme Prodrug Therapy (ADEPT) for the treatment of cancer. In common with many protein therapeutics, glucarpidase has a relatively short half-life in serum and, due to the need for the repeated cycles of the ADEPT, its bioavailability may be further diminished by neutralizing antibodies produced by patients. PEGylation and fusion with human serum albumin (HSA) are two approaches that are commonly employed to increase the residency time of protein therapeutics in blood, and also to increase the half-lives of the proteins in vivo. To address this stability and the immunogenicity problems, 'biobetter' glucarpidase variants, mono-PEGylated glucarpidase, and HSA fused glucarpidase by genetic fusion with albumin, were produced. Biochemical and bioactivity analyses, including anti-proliferation, bioassays, circular dichroism, and in vitro stability using human blood serum and immunoassays, demonstrated that the functional activities of the designed glucarpidase conjugates were maintained. The immunotoxicity studies indicated that the PEGylated glucarpidase did not significantly induce T-cell proliferation, suggesting that glucarpidase epitopes were masked by the PEG moiety. However, free glucarpidase and HSA-glucarpidase significantly increased T-cell proliferation compared with the negative control. In the latter case, this might be due to the type of expression system used or due to trace impurities associated with the highly purified (99.99%) recombinant HSA-glucarpidase. Both PEGylated glucarpidase and HAS-glucarpidase exhibit more stability in human serum and were more resistant to key human proteases relative to native glucarpidase. To our knowledge, this study is the first to report stable and less immunogenic glucarpidase variants produced by PEGylation and fusion with HSA. The results suggest that they may have better efficacy in drug detoxification and ADEPT, thereby improving this cancer treatment strategy.


Subject(s)
Antibodies/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Methotrexate/pharmacology , Polyethylene Glycols/administration & dosage , Prodrugs/administration & dosage , Serum Albumin, Human/administration & dosage , gamma-Glutamyl Hydrolase/administration & dosage , Antibodies/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Therapy , Humans , Hydrolysis , Leukocytes, Mononuclear/drug effects , Neoplasms/drug therapy , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Serum Albumin, Human/chemistry , Serum Albumin, Human/genetics , T-Lymphocytes/drug effects , gamma-Glutamyl Hydrolase/chemistry
12.
PLoS One ; 13(4): e0196254, 2018.
Article in English | MEDLINE | ID: mdl-29698433

ABSTRACT

Repeated cycles of antibody-directed enzyme pro-drug therapy (ADEPT) and the use of glucarpidase in the detoxification of cytotoxic methotrexate (MTX) are highly desirable during cancer therapy but are hampered by the induced human antibody response to glucarpidase. Novel variants of glucarpidase (formal name: carboxypeptidase G2, CPG2) with epitopes not recognized by the immune system are likely to allow repeated cycles of ADEPT for effective cancer therapy. Towards this aim, over two thousand soil samples were collected and screened for folate hydrolyzing bacteria using folate as the sole carbon source. The work led to the isolation and the characterization of three new glucarpidase producing strains, which were designated as: Pseudomonas lubricans strain SF168, Stenotrophomonas sp SA and Xenophilus azovorans SN213. The CPG2 genes of Xenophilus azovorans SN213 (named Xen CPG2) and Stenotrophomonas sp SA (named Sten CPG2) were cloned and molecularly characterized. Both Xen CPG2 and Sten CPG2 share very close amino acid sequences (99%); we therefore, focused on the study of Xen CPG2. Finally, we demonstrated that a polyclonal antibody raised against our new CPG2, Xen CPG2, does not react with the CPG2 from Pseudomonas sp. strain RS-16 (Ps CPG2) that are currently in clinical use. The two enzymes, therefore could potentially be used consecutively in the ADEPT protocol to minimize the effect of the human antibody response that hampers current treatment with Ps CPG2. The identified novel CPG2 in this study will, therefore, pave the way for safer antibody directed enzyme pro-drug therapy for cancer treatment.


Subject(s)
Antibodies/chemistry , Methotrexate/pharmacology , Neoplasms/drug therapy , gamma-Glutamyl Hydrolase/chemistry , Carbon/chemistry , Circular Dichroism , Cloning, Molecular , Folic Acid/chemistry , Humans , Hydrolysis , Immune System , Mass Spectrometry , Neoplasms/immunology , Prodrugs/therapeutic use , Pseudomonas/enzymology , Recombinant Proteins/chemistry , Stenotrophomonas/enzymology , Zinc/chemistry
13.
ACS Chem Biol ; 9(3): 802-11, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24405416

ABSTRACT

The inhibition of p53-MDM2 interaction is a promising new approach to non-genotoxic cancer treatment. A potential application for drugs blocking the p53-MDM2 interaction is acute myeloid leukemia (AML) due to the occurrence of wild type p53 (wt p53) in the majority of patients. Although there are very promising preclinical results of several p53-MDM2 antagonists in early development, none of the compounds have yet proven the utility as a next generation anticancer agent. Herein we report the design, synthesis and optimization of YH239-EE (ethyl ester of the free carboxylic acid compound YH239), a potent p53-MDM2 antagonizing and apoptosis-inducing agent characterized by a number of leukemia cell lines as well as patient-derived AML blast samples. The structural basis of the interaction between MDM2 (the p53 receptor) and YH239 is elucidated by a co-crystal structure. YH239-EE acts as a prodrug and is the most potent compound that induces apoptosis in AML cells and patient samples. The observed superior activity compared to reference compounds provides the preclinical basis for further investigation and progression of YH239-EE.


Subject(s)
Antineoplastic Agents , Drug Discovery , Indoles , Leukemia, Myeloid, Acute/drug therapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Humans , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Models, Molecular , Molecular Structure , Protein Binding , Proto-Oncogene Proteins c-mdm2/chemistry , Structure-Activity Relationship
14.
Biodegradation ; 21(6): 903-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20401686

ABSTRACT

Five malathion-degrading bacterial strains were enriched and isolated from soil samples collected from different agricultural sites in Cairo, Egypt. Malathion was used as a sole source of carbon (50 mg/l) to enumerate malathion degraders, which were designated as IS1, IS2, IS3, IS4, and IS5. They were identified, based on their morphological and biochemical characteristics, as Pseudomonas sp., Pseudomonas putida, Micrococcus lylae, Pseudomonas aureofaciens, and Acetobacter liquefaciens, respectively. IS1 and IS2, which showed the highest degrading activity, were selected for further identification by partial sequence analysis of their 16S rRNA genes. The 16S rRNA gene of IS1 shared 99% similarity with that of Alphaprotoebacterium BAL284, while IS2 scored 100% similarity with that of Pseudomonas putida 32zhy. Malathion residues almost completely disappeared within 6 days of incubation in IS2 liquid cultures. LC/ESI-MS analysis confirmed the degradation of malathion to malathion monocarboxylic and dicarboxylic acids, which formed as a result of carboxylesterase activity. A carboxylesterase gene (CE) was amplified from the IS2 genome by using specifically designed PCR primers. The sequence analysis showed a significant similarity to a known CE gene in different Pseudomonas sp. We report here the isolation of a new malathion-degrading bacteria from soils in Egypt that may be very well adapted to the climatic and environmental conditions of the country. We also report the partial cloning of a new CE gene. Due to their high biodegradation activity, the bacteria isolated from this work merit further study as potential biological agents for the remediation of soil, water, or crops contaminated with the pesticide malathion.


Subject(s)
Bacteria/genetics , Bacteria/isolation & purification , Carboxylesterase/genetics , Genes, Bacterial/genetics , Malathion/metabolism , Sequence Analysis, DNA , Soil Microbiology , Amino Acid Sequence , Bacteria/enzymology , Biodegradation, Environmental , Carboxylesterase/chemistry , Chromatography, Gas , Cloning, Molecular , Electrophoresis, Agar Gel , Malathion/chemistry , Mass Spectrometry , Molecular Sequence Data , Pesticides/metabolism , Polymerase Chain Reaction , Pseudomonas putida/growth & development , Pseudomonas putida/metabolism , RNA, Ribosomal/genetics , Sequence Alignment
15.
Protein J ; 29(1): 44-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20033268

ABSTRACT

The aim of this study was to evaluate the possibility that synthetic forms of methionine-free alpha-casein and methionine-limited alpha casein could be produced by recombinant means to form the basis for developing an industrial-scale process for the provision of a foodstuff suitable for patients with homocystinuria due to cystathionine beta-synthase (CBS) deficiency. As a first step, two forms of alpha casein gene, encoding methionine-free alpha casein (Fcas) or a methionine-limited alpha casein (Mcas), were synthesised and expressed in Escherichia coli. Using the overexpression vector pET28a, both genes were highly expressed in E. coli in soluble form as well as in inclusion bodies. The two recombinant proteins were purified by the one step methods using the fused His-tag and the Ni(2+)column and validated by Western blot analysis. This work paves the way for industrial-scale production of proteins suitable for patients with homocystinuria due to CBS deficiency.


Subject(s)
Caseins/metabolism , Cystathionine beta-Synthase/deficiency , Homocystinuria/enzymology , Methionine/analysis , Protein Engineering , Caseins/analysis , Caseins/genetics , Caseins/isolation & purification , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Homocystinuria/diet therapy , Humans , Methionine/genetics , Methionine/metabolism , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
16.
Protein J ; 28(9-10): 435-42, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19911261

ABSTRACT

Glucarpidase (former name: carboxypeptidase G2, or CPG2) is a bacterial enzyme that is widely used in detoxification of the cytotoxic drug, methotrexate, and in Antibody Directed Enzyme Prodrug Therapy for cancer treatment. The glucarpidase gene of Pseudomonas sp. strain RS-16 was previously cloned in E coli, but expresses at a level that is approximately 100-fold lower than in the native strain. In this study, a synthetic gene coding for glucarpidase was codon-optimised and synthesized for maximum expression in E. coli using the vector pET28a. Our work indicated that the enzyme was expressed to ~60% of the total host protein and that purification of the recombinant His-tagged protein could be achieved in a single step by Ni(2+) charged column chromatography. The synthetic recombinant glucarpidase expressed within this system was biologically active and zinc dependant. Our study showed that Mg(2+) as well as Mn(2+) ions inhibit the activity of the recombinant enzyme.


Subject(s)
Escherichia coli/genetics , gamma-Glutamyl Hydrolase/genetics , gamma-Glutamyl Hydrolase/metabolism , Amino Acid Sequence , Cloning, Molecular , Gene Expression Regulation, Bacterial , Genes, Bacterial , Inclusion Bodies/chemistry , Molecular Sequence Data , Protein Folding , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Zinc/metabolism , gamma-Glutamyl Hydrolase/isolation & purification
17.
J Biol Chem ; 279(2): 1297-303, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14559915

ABSTRACT

The previously determined crystal structure of the superantigen staphylococcal enterotoxin C2 (SEC2) showed binding of a single zinc ion located between the N- and C-terminal domains. Here we present the crystal structure of SEC2 determined to 2.0 A resolution in the presence of additional zinc. The structure revealed the presence of a secondary zinc-binding site close to the major histocompatibility complex (MHC)-binding site of the toxin and some 28 A away from the primary zinc-binding site of the toxin found in previous studies. T cell stimulation assays showed that varying the concentration of zinc ions present affected the activity of the toxin and we observed that high zinc concentrations considerably inhibited T cell responses. This indicates that SEC2 may have multiple modes of interaction with the immune system that are dependent on serum zinc levels. The potential role of the secondary zinc-binding site and that of the primary one in the formation of the TCR.SEC2.MHC complex are considered, and the possibility that zinc may regulate the activity of SEC2 as a toxin facilitating different T cell responses is discussed.


Subject(s)
Enterotoxins/chemistry , Zinc/chemistry , Animals , Antigens, Bacterial/chemistry , Binding Sites , CHO Cells , Cell Division , Cricetinae , Crystallography, X-Ray , Dimerization , Humans , Models, Molecular , T-Lymphocytes/metabolism , Transfection , X-Ray Diffraction
18.
Microbiology (Reading) ; 143 ( Pt 10): 3287-3294, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9353929

ABSTRACT

An Escherichia coli clone was detected in a Clostridium butyricum NCIMB 7423 plasmid library capable of degrading soluble amylose. Deletion subcloning of its recombinant plasmid indicated that the gene(s) responsible for amylose degradation was localized on a 1.8 kb NspHI-Scal fragment. This region was sequenced in its entirety and shown to encompass a large ORF capable of encoding a protein with a calculated molecular mass of 57,184 Da. Although the deduced amino acid sequence showed only weak similarity with known amylases, significant sequences identity was apparent with the 4-alpha-glucano-transferase enzymes of Streptococcus pneumoniae (46.9%), potato (42.9%) and E. coli (16.2%). The clostridial gene (designated maIQ) was followed by a second ORF which, through its homology to the equivalent enzymes of E. coli and S. pneumoniae, was deduced to encode maltodextrin phosphorylase (MaIP). The translation stop codon of MaIQ overlapped the translation start codon of the putative maIP gene, suggesting that the two genes may be both transcriptionally and translationally coupled. 4-alpha-Glucanotransferase catalyses a disproportionation reaction in which single or multiple glucose units from oligosaccharides are transferred to the 4-hydroxyl group of acceptor sugars. Characterization of the recombinant C. butyricum enzyme demonstrated that glucose, maltose and maltotriose could act as acceptor, whereas of the three only maltotriose could act as donor. The enzyme therefore shares properties with the E. coli MaIQ protein, but differs significantly from the glucanotransferase of Thermotoga maritima, which is unable to use maltotriose as donor or glucose as acceptor. Physiologically, the concerted action of 4-alpha-glucanotransferase and maltodextrin phosphorylase provides C. butyricum with a mechanism of utilizing amylose/maltodextrins with little drain on cellular ATP reserves.


Subject(s)
Clostridium/enzymology , Clostridium/genetics , Genes, Bacterial , Glycogen Debranching Enzyme System/genetics , Amino Acid Sequence , Amylose/metabolism , Base Sequence , Cloning, Molecular , DNA, Bacterial/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Glucosyltransferases/genetics , Glycogen Debranching Enzyme System/biosynthesis , Glycogen Debranching Enzyme System/metabolism , Molecular Sequence Data , Molecular Weight , Open Reading Frames , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...