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










Publication year range
1.
Cell Biochem Biophys ; 67(1): 161-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23695783

ABSTRACT

The ubiquitin pathway regulates diverse functions including protein localization and stability. The complexity of the pathway involving nearly 40 identified E2 conjugating enzymes and over 600 E3 ligases raises the issue of specificity. With the E2s and E3s fitting into a limited number of classes based on bioinformatics, structures, and proven activities, there is not a clear picture as to what would determine which E2/E3 enzyme pair would be functional. There have been many reports of limited E2/E3 activity profiling with a small number of E2s and E3s. We have expanded on this to investigate the activity of ubiquitin E2s covering the majority of the reported classes/families in concert with a number of E3s implicated in a variety of diseases. Using an ELISA-based assay we screened 10 E3 ligases against a panel of 11 E2s to determine which E2/E3 pairs exhibited E3 autoubiquitylation activity. In addition, the ubiquitin chain linkage preference by certain E2/E3 pairs was investigated. Finally, substrate ubiquitylation was assayed for the E3 ligase MuRF1 using various E2/MuRF1 pairs. These studies demonstrate the utility of identifying the correct E2/E3 pair to monitor specific substrate ubiquitylation.


Subject(s)
Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Substrate Specificity , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
Biochim Biophys Acta ; 1823(11): 2094-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22721718

ABSTRACT

The ubiquitin proteasome pathway controls the cellular degradation of ~80-90% of the proteome in a highly regulated manner. In this pathway, E3 ligases are responsible for the conjugation of ubiquitin to protein substrates which can lead to their destruction by the 26S proteasome. Aberrant E3 ligases have been implicated in several diseases and are widely recognized as attractive targets for drug discovery. As researchers continue to characterize E3 ligases, additional associations with various disease states are being exposed. The availability of assays that allow rapid analysis of E3 ligase activity is paramount to both biochemical studies and drug discovery efforts aimed at E3 ligases. To address this need, we have developed a homogenous assay for monitoring ubiquitin chain formation using Tandem Ubiquitin Binding Entities (TUBEs). TUBEs bind selectively to polyubiquitin chains versus mono-ubiquitin thus enabling the detection of polyubiquitin chains in the presence of mono-ubiquitin. This assay reports on the proximity between the protein substrate and TUBEs as a result of polyubiquitin chain formation by an E3 ligase. This homogenous assay is a step forward in streamlining an approach for characterizing and quantitating E3 ligase activity in a rapid and cost effective manner. This article is part of a Special Issue entitled: Ubiquitin Drug Discovery and Diagnostics.


Subject(s)
Muscle Proteins/metabolism , Polyubiquitin/metabolism , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , High-Throughput Screening Assays , Humans , Protein Binding , Tandem Repeat Sequences , Tripartite Motif Proteins , Ubiquitination
3.
Biochim Biophys Acta ; 1823(11): 2014-21, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22610084

ABSTRACT

The proteasome inhibitor bortezomib remains the only ubiquitin pathway effector to become a drug (VELCADE®) and has become a successful treatment for hematological malignancies. While producing a global cellular effect, proteasome inhibitors have not triggered the catastrophe articulated initially in terms such as "buildup of cellular garbage". Proteasome inhibitors, in fact, do have a therapeutic window, although in the case of the prototype bortezomib it is small owing to peripheral neuropathy, myelosuppression and, as recently reported, cardiotoxicity [1]. Currently, several second-generation molecules are undergoing clinical evaluation to increase this window. An alternative strategy is to target ubiquitin pathway enzymes acting at non-proteasomal sites-E1, E2, and E3, associated with ubiquitin conjugation, and deubiquitylating enzymes ("DUBs")-that act locally on selected targets rather than on the whole cell. Inhibitors (or activators, in some cases) of these enzymes should be developable as selective antitumor agents with toxicity profiles superior to that of bortezomib. Various therapeutic hypotheses follow from known cellular mechanisms of these target enzymes; most hypotheses relate to cancer, reminiscent of the FDA-approved protein kinase inhibitors now marketed. Since ubiquitin tagging controls the cellular content, activity, or compartmentation of proteins associated with disease, inhibitors or activators of ubiquitin conjugation or deconjugation are predicted to have an impact on disease. For practical and empirical reasons, inhibitors of ubiquitin pathway enzymes have been the favored therapeutic avenue. In approximately the time that has elapsed since the approval of bortezomib in 2003, there has been some progress in developing potential anticancer drugs that target various ubiquitin pathway enzymes. An E1 inhibitor and inhibitors of E3 are now in clinical trial, with some objective responses reported. Appropriate assays and/or rational design may uncover improved inhibitors of these enzymes, as well as E2 and DUBs, for further development. Presently, it should become clear whether one or both of the two general strategies for ubiquitin-based drug discovery will lead to truly superior new medicines for cancer and other diseases. This article is part of a Special Issue entitled: Ubiquitin Drug Discovery and Diagnostics.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Protease Inhibitors/therapeutic use , Proteasome Endopeptidase Complex/chemistry , Ubiquitin/metabolism , Animals , Clinical Trials as Topic , Humans
4.
ACS Med Chem Lett ; 3(10): 789-92, 2012 Oct 11.
Article in English | MEDLINE | ID: mdl-24900381

ABSTRACT

Inhibitors of the cancer-related cysteine isopeptidase human ubiquitin-specific proteases 7 (USP7) and 47 (USP47) are considered to have potential as cancer therapeutics, owing to their ability to stabilize the tumor suppressor p53 and to decrease DNA polymerase ß (Polß), both of which are potential anticancer effects. A new class of dual small molecule inhibitors of these enzymes has been discovered. Compound 1, a selective inhibitor of USP7 and USP47 with moderate potency, demonstrates inhibition of USP7 in cells and induces elevated p53 and apoptosis in cancer cell lines. Compound 1 has been shown to demonstrate modest activity in human xenograft multiple myeloma and B-cell leukemia in vivo models. This activity may be the result of dual inhibition of USP7 and USP47. To address issues regarding potency and developability, analogues of compound 1 have been synthesized and tested, leading to improvements in potency, solubility, and metabolic reactivity profile. Further optimization is expected to yield preclinical candidates and, ultimately, clinical candidates for the treatment of multiple myeloma, prostate cancer, and other cancers.

5.
Cell Biochem Biophys ; 60(1-2): 113-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21448668

ABSTRACT

Progressive muscle wasting, also known as myopathy or muscle atrophy is a debilitating and life-threatening disorder. Myopathy is a pathological condition of many diseases including cancer, diabetes, COPD, and AIDS and is a natural consequence of inactivity and aging (sarcopenia). Muscle atrophy occurs when there is a net loss of muscle mass resulting in a change in the balance between protein synthesis and protein degradation. The ubiquitin pathway and specific ubiquitin pathway enzymes have been directly implicated in the progression of atrophy. The ubiquitin E3 ligase Muscle-specific RING Finger E3 ligase (MuRF1) is upregulated and increases protein degradation and muscle wasting in numerous muscle atrophy models. The inhibition of MuRF1 could be a novel mechanism to prevent or reverse muscle wasting associated with various pathologies. We screened a small molecule library for inhibitors to MuRF1 activity and identified P013222, an inhibitor of MuRF1 autoubiquitylation. Further, P013222 was shown to inhibit MuRF1-dependent substrate ubiquitylation, and was active in inhibiting MuRF1 in a cellular atrophy model. Thus MuRF1 can be targeted in a specific manner and produce positive results in cellular atrophy models.


Subject(s)
Enzyme Inhibitors/pharmacology , Muscle Proteins/antagonists & inhibitors , Muscular Atrophy/prevention & control , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , Biocatalysis/drug effects , Blotting, Western , Cell Line , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Glucocorticoids/pharmacology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Small Molecule Libraries , Substrate Specificity , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
6.
Assay Drug Dev Technol ; 9(2): 165-73, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21133675

ABSTRACT

The reversible conjugation of ubiquitin and ubiquitin-like (UbL) proteins to protein substrates plays a critical role in the regulation of many cellular pathways. The removal of ubiquitin from target proteins is performed by ubiquitin proteases also known as deubiquitylases (DUBs). Owing to their substrate specificity and the central role ubiquitylation plays in cell signaling pathways, DUB are attractive targets for therapeutic development. The development of DUB inhibitors requires assays that are amenable to high-throughput screening and provide rapid assessment of inhibitor selectivity. Determination of inhibitor selectivity at an early stage of drug discovery will reduce drug failure in the clinic as well as reduce overall drug development costs. We have developed two novel assays, UbL-Enterokinase light chain and UbL-Granzyme B, for quantifying ubiquitin and UbL protease activity. In our quest to discover and characterize novel chemical entities, we have combined these assays with a previously developed assay in a multiplex format. This multiplex format allows for the detection of three distinct protease activities simultaneously, in a single well. We have demonstrated that the multiplex format is able to distinguish between selective and nonselective protease inhibitors. Specifically, we have used this assay format to characterize P022077, a selective ubiquitin-specific protease 7 inhibitor discovered at Progenra.


Subject(s)
Fluorescent Dyes/analysis , Protease Inhibitors/chemistry , Ubiquitin/antagonists & inhibitors , Ubiquitins/antagonists & inhibitors , Clinical Laboratory Techniques , Fluorescence , Fluorescent Dyes/metabolism , Pichia , Protease Inhibitors/analysis , Protease Inhibitors/metabolism , Substrate Specificity/physiology , Ubiquitin/metabolism , Ubiquitins/metabolism
7.
J Biomol Screen ; 15(10): 1220-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20864734

ABSTRACT

The ubiquitin-proteasome system is central to the regulation of numerous cellular events, and dysregulation may lead to disease pathogenesis. E3 ubiquitin ligases typically function in concert with E1 and E2 enzymes to recruit specific substrates, thereby coordinating their ubiquitylation and subsequent proteasomal degradation or cellular activity. E3 ligases have been implicated in a wide range of pathologies, and monitoring their activity in a rapid and cost-effective manner would be advantageous in drug discovery. The relative lack of high-throughput screening (HTS)-compliant E3 ligase assays has significantly hindered the discovery of E3 inhibitors. Herein, the authors describe a novel HTS-compliant E3 ligase assay platform that takes advantage of a ubiquitin binding domain's inherent affinity for polyubiquitin chains, permitting the analysis of ubiquitin chain formation in an E3 ligase-dependent manner. This assay has been used successfully with members of both the RING and HECT families, demonstrating the platform's broad utility for analyzing a wide range of E3 ligases. The utility of the assay platform is demonstrated by the identification of inhibitors of the E3 ligase CARP2. As the number of E3 ligases associated with various disease states increases, the ability to quantitate the activity of these enzymes in an expeditious manner becomes imperative in drug discovery.


Subject(s)
DNA Repair Enzymes/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , High-Throughput Screening Assays/methods , Nerve Tissue Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Drug Discovery , Humans , Luminescence , Nerve Tissue Proteins/metabolism , Polyubiquitin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
8.
Biochem Soc Trans ; 38(Pt 1): 132-6, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20074047

ABSTRACT

Dysregulation of the UPS (ubiquitin-proteasome system) has been implicated in a wide range of pathologies including cancer, neurodegeneration and viral infection. Inhibiting the proteasome has been shown to be an effective therapeutic strategy in humans; however, toxicity with this target remains high. E3s (Ub-protein ligases) represent an alternative attractive therapeutic target in the UPS. In this paper, we will discuss current platforms that report on E3 ligase activity and can detect E3 inhibitors, and underline the advantages and disadvantages of each approach.


Subject(s)
Protease Inhibitors/metabolism , Proteasome Inhibitors , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin/metabolism , Biological Assay/methods , Humans , Immune System Diseases/drug therapy , Immune System Diseases/enzymology , Neoplasms/drug therapy , Neoplasms/enzymology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism
9.
Methods Mol Biol ; 497: 269-81, 2009.
Article in English | MEDLINE | ID: mdl-19107424

ABSTRACT

In this chapter we describe a novel, sensitive, homogenous high throughput reporter-based in vitro assay for SUMO protease activity developed by Progenra, Inc. A reporter construct was created by fusing His(6)-tagged small ubiquitin-like modifier (SUMO) to the amino terminus of the reporter enzyme phospholipase A(2) (PLA(2)). Following cleavage by a member of the sentrin specific proteases (SENPs), free PLA(2) is able to turn over its substrate, resulting in the release of a fluorescent product which is readily quantifiable using a fluorimeter or a fluorescence plate reader. The utility of this SUMO-CHOP-Reporter assay platform is demonstrated by its ability to determine K(m) values and to characterize inhibitors of SUMO proteases.


Subject(s)
Biosensing Techniques/methods , Peptide Hydrolases/isolation & purification , Peptide Hydrolases/metabolism , SUMO-1 Protein/metabolism , Catalysis , Clinical Laboratory Techniques , Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Humans , Kinetics , Peptide Hydrolases/chemistry , Phospholipases A2/metabolism , Protein Processing, Post-Translational , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sensitivity and Specificity , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factor CHOP/chemistry , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/physiology
10.
Biochem Soc Trans ; 36(Pt 5): 828-32, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18793145

ABSTRACT

Dysregulation of the UPS (ubiquitin-proteasome system) has been implicated in a wide range of pathologies including cancer, neurodegeneration and viral infection. Inhibiting the proteasome has been shown to be an effective therapeutic strategy in humans; yet toxicity with this target remains high. DUBs (deubiquitinating enzymes) represent an alternative target in the UPS with low predicted toxicity. Currently, there are no DUB inhibitors that have been used clinically. To address this situation, Progenra has developed a novel assay to measure the proteolytic cleavage of Ub (ubiquitin) or UBL (Ub-like protein) conjugates such as SUMO (small Ub-related modifier), NEDD8 (neural-precursor-cell-expressed, developmentally down-regulated 8) or ISG15 (interferon-stimulated gene 15) by isopeptidases. In this review, current platforms for detecting DUB inhibitors are discussed and the advantages and disadvantages of the approaches are underlined.


Subject(s)
Biological Assay/methods , Endopeptidases/metabolism , Enzyme Inhibitors/metabolism , Ubiquitin/metabolism , Disease , Endopeptidases/genetics , Enzyme Inhibitors/therapeutic use , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Phospholipases A2/genetics , Phospholipases A2/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/genetics
11.
Protein Sci ; 17(6): 1035-43, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18424514

ABSTRACT

Conjugation or deconjugation of ubiquitin (Ub) or ubiquitin-like proteins (UBLs) to or from cellular proteins is a multifaceted and universal means of regulating cellular physiology, controlling the lifetime, localization, and activity of many critical proteins. Deconjugation of Ub or UBL from proteins is performed by a class of proteases called isopeptidases. Herein is described a readily quantifiable novel isopeptidase assay platform consisting of Ub or UBL fused to the reporter enzyme phospholipase A(2) (PLA(2)). Isopeptidase activity releases PLA(2), which cleaves its substrate, generating a signal that is linear with deubiquitylase (DUB) concentration and is able to discriminate DUB, deSUMOylase, deNEDDylase, and deISGylase activities. The power and sensitivity of the UBL-PLA(2) assay are demonstrated by its ability to differentiate the contrasting deISGylase and DUB activities of two coronavirus proteases: severe acute respiratory syndrome papain-like protease (SARS-CoV PLpro) and NL63 CoV papain-like protease 2 (PLP2). Furthermore, direct comparisons with the current Ub-7-amino-4-methylcoumarin (Ub-AMC) assay demonstrated that the Ub-PLA(2) assay is an effective tool for characterizing modulators of isopeptidase activity. This observation was expanded by profiling the inhibitory activity of the nonselective isopeptidase inhibitor NSC 632839 against DUBs and deSUMOylases. Taken together, these studies illustrate the utility of the reporter-based approach to measuring isopeptidase activity.


Subject(s)
Endopeptidases/metabolism , Ubiquitin/metabolism , Animals , Endopeptidases/isolation & purification , Mice , Protease Inhibitors/pharmacology
12.
Future Oncol ; 3(2): 191-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17381419

ABSTRACT

Tagging proteins with mono- or poly-ubiquitin is now recognized as a multifaceted and universal means of regulating cell growth and physiology. It does so by controlling the cellular lifetime of nearly all eukaryotic proteins and the cellular localization of many critical proteins. Enzymes of the ubiquitin pathway add (ligases) or remove (deubiquitinases [DUBs]) ubiquitin tags to or from their target proteins in a selective fashion. Similarly to the kinases and their corresponding phosphatases, ubiquitin ligases and DUBs have become actively studied molecular oncology targets for drug discovery. Approximately 79 functional DUBs exist in the human proteome, suggesting that selective intervention is a reasonable therapeutic objective, with the goal of downregulating or ablating oncogene products or, alternatively, upregulating or sparing tumor suppressors. In the following review, this fascinating class of regulatory enzymes will be described, and specific examples of DUBs that are viable targets for anticancer therapy will be considered.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Ubiquitin/drug effects , Ubiquitin/metabolism , Humans , NEDD8 Protein , Proteasome Endopeptidase Complex/metabolism , Small Ubiquitin-Related Modifier Proteins/drug effects , Ubiquitin Thiolesterase/drug effects , Ubiquitins/drug effects
13.
Anal Biochem ; 350(2): 214-21, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16356462

ABSTRACT

The ubiquitin-proteasome pathway is the major nonlysosomal proteolytic system in eukaryotic cells responsible for regulating the level of many key regulatory molecules within the cells. Modification of cellular proteins by ubiquitin and ubiquitin-like proteins, such as small ubiquitin-like modifying protein (SUMO), plays an essential role in a number of biological schemes, and ubiquitin pathway enzymes have become important therapeutic targets. Ubiquitination is a dynamic reversible process; a multitude of ubiquitin ligases and deubiquitinases (DUBs) are responsible for the wide-ranging influence of this pathway as well as its selectivity. The DUB enzymes serve to maintain adequate pools of free ubiquitin and regulate the ubiquitination status of cellular proteins. Using SUMO fusions, a novel assay system, based on poliovirus RNA-dependent RNA polymerase activity, is described here. The method simplifies the isopeptidase assay and facilitates high-throughput analysis of these enzymes. The principle of the assay is the dependence of the viral polymerase on a free N terminus for activity; accordingly, the polymerase is inactive when fused at its N terminus to SUMO or any other ubiquitin-like protein. The assay is sensitive, reproducible, and adaptable to a high-throughput format for use in screens for inhibitors/activators of clinically relevant SUMO proteases and deubiquitinases.


Subject(s)
Carbon-Nitrogen Lyases/analysis , DNA-Directed RNA Polymerases/metabolism , Poliovirus/enzymology , SUMO-1 Protein/analysis , Ubiquitins/analysis , Recombinant Fusion Proteins/metabolism
14.
J Struct Funct Genomics ; 6(2-3): 103-11, 2005.
Article in English | MEDLINE | ID: mdl-16211506

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV) membrane protein and 5-lipoxygenase-activating protein (FLAP) are among a large number of membrane proteins that are poorly expressed when traditional expression systems and methods are employed. Therefore to efficiently express difficult membrane proteins, molecular biologists will have to develop novel or innovative expression systems. To this end, we have expressed the SARS-CoV M and FLAP proteins in Escherichia coli by utilizing a novel gene fusion expression system that takes advantage of the natural chaperoning properties of the SUMO (small ubiquitin-related modifier) tag. These chaperoning properties facilitate proper protein folding, which enhances the solubility and biological activity of the purified protein. In addition to these advantages, we found that SUMO Protease 1, can cleave the SUMO fusion high specificity to generate native protein. Herein, we demonstrate that the expression of FLAP and SARS-CoV membrane proteins are greatly enhanced by SUMO fusions in E. coli.


Subject(s)
Carrier Proteins/isolation & purification , Escherichia coli/chemistry , Membrane Proteins/isolation & purification , SUMO-1 Protein/metabolism , Viral Matrix Proteins/isolation & purification , 5-Lipoxygenase-Activating Proteins , Blotting, Western , Carrier Proteins/metabolism , Coronavirus M Proteins , DNA Primers , Databases, Protein , Genetic Vectors/genetics , Membrane Proteins/metabolism , Protein Folding , Viral Matrix Proteins/metabolism
15.
J Med Chem ; 48(18): 5644-7, 2005 Sep 08.
Article in English | MEDLINE | ID: mdl-16134930

ABSTRACT

Inhibitors of human methionine aminopeptidase type 2 (hMetAP2) are of interest as potential treatments for cancer. A new class of small molecule reversible inhibitors of hMetAP2 was discovered and optimized, the 4-aryl-1,2,3-triazoles. Compound 24, a potent inhibitor of cobalt-activated hMetAP2, also inhibits human and mouse endothelial cell growth. Using a mouse matrigel model, this reversible hMetAP2 inhibitor was also shown to inhibit angiogenesis in vivo.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Angiogenesis Inhibitors/chemical synthesis , Metalloendopeptidases/antagonists & inhibitors , Triazoles/chemical synthesis , Aminopeptidases/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Binding Sites , Biological Availability , Cell Proliferation/drug effects , Cells, Cultured , Cobalt/metabolism , Collagen , Crystallography, X-Ray , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelium, Vascular/cytology , Enzyme Activation , Humans , Laminin , Metalloendopeptidases/chemistry , Mice , Models, Molecular , Molecular Structure , Proteoglycans , Rats , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/pharmacology
16.
Protein Expr Purif ; 43(1): 1-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16084395

ABSTRACT

The demands of structural and functional genomics for large quantities of soluble, properly folded proteins in heterologous hosts have been aided by advancements in the field of protein production and purification. Escherichia coli, the preferred host for recombinant protein expression, presents many challenges which must be surmounted in order to over-express heterologous proteins. These challenges include the proteolytic degradation of target proteins, protein misfolding, poor solubility, and the necessity for good purification methodologies. Gene fusion technologies have been able to improve heterologous expression by overcoming many of these challenges. The ability of gene fusions to improve expression, solubility, purification, and decrease proteolytic degradation will be discussed in this review. The main disadvantage, cleaving the protein fusion, will also be addressed. Focus will be given to the newly described SUMO fusion system and the improvements that this technology has advanced over traditional gene fusion systems.


Subject(s)
Gene Fusion/methods , Recombinant Fusion Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Escherichia coli/metabolism , Gene Expression , Protein Folding
17.
Protein Expr Purif ; 42(1): 100-10, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15939295

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV) proteins belong to a large group of proteins that is difficult to express in traditional expression systems. The ability to express and purify SARS-CoV proteins in large quantities is critical for basic research and for development of pharmaceutical agents. The work reported here demonstrates: (1) fusion of SUMO (small ubiquitin-related modifier), a 100 amino acid polypeptide, to the N-termini of SARS-CoV proteins dramatically enhances expression in Escherichia coli cells and (2) 6x His-tagged SUMO-fusions facilitate rapid purification of the viral proteins on a large scale. We have exploited the natural chaperoning properties of SUMO to develop an expression system suitable for proteins that cannot be expressed by traditional methodologies. A unique feature of the system is the SUMO tag, which enhances expression, facilitates purification, and can be efficiently cleaved by a SUMO-specific protease to generate native protein with a desired N-terminus. We have purified various SARS-CoV proteins under either native or denaturing conditions. These purified proteins have been used to generate highly specific polyclonal antibodies. Our study suggests that the SUMO-fusion technology will be useful for enhancing expression and purification of the viral proteins for structural and functional studies as well as for therapeutic uses.


Subject(s)
Gene Expression/genetics , Recombinant Fusion Proteins/biosynthesis , Severe acute respiratory syndrome-related coronavirus/genetics , Small Ubiquitin-Related Modifier Proteins/genetics , Viral Proteins/genetics , Coronavirus 3C Proteases , Coronavirus Nucleocapsid Proteins , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/isolation & purification , Escherichia coli/genetics , Genetic Vectors/genetics , Histidine/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Glycoproteins/isolation & purification , Nucleocapsid Proteins/biosynthesis , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/isolation & purification , Peptide Hydrolases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/isolation & purification , Viral Proteins/isolation & purification , Viral Proteins/metabolism
18.
J Struct Funct Genomics ; 5(1-2): 75-86, 2004.
Article in English | MEDLINE | ID: mdl-15263846

ABSTRACT

SUMO (small ubiquitin-related modifier) modulates protein structure and function by covalently binding to the lysine side chains of the target proteins. Yeast cells contain two SUMO proteases, Ulp1 and Ulp2, that cleave sumoylated proteins in the cell. Ulp1 (SUMO protease 1) processes the SUMO precursor to its mature form and also de-conjugates SUMO from side chain lysines of target proteins. Here we demonstrate that attachment of SUMO to the N-terminus of under-expressed proteins dramatically enhances their expression in E. coli. SUMO protease 1 was able to cleave a variety of SUMO fusions robustly and with impeccable specificity. Purified recombinant SUMO-GFPs were efficiently cleaved when any amino acid, except proline, was in the+1 position of the cleavage site. The enzyme was active over a broad range of buffer and temperature conditions. Purification of certain recombinant proteins is accomplished by production of Ub-fusions from which Ub can be subsequently removed by de-ubiquitinating enzymes (DUBs). However, DUBs are unstable enzymes that are difficult to produce and inexpensive DUBs are not available commercially. Our findings demonstrate that SUMO protease 1/SUMO-fusion system may be preferable to DUB/Ub-fusion. Enhanced expression and solubility of proteins fused to SUMO combined with broad specificity and highly efficient cleavage properties of the SUMO protease 1 indicates that SUMO-fusion technology will become a useful tool in purification of proteins and peptides.


Subject(s)
Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Small Ubiquitin-Related Modifier Proteins/biosynthesis , Small Ubiquitin-Related Modifier Proteins/genetics , Base Sequence , Catalytic Domain , Cloning, Molecular , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , DNA Primers/genetics , Endopeptidases/chemistry , Endopeptidases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Hydrogen-Ion Concentration , Molecular Sequence Data , Polymerase Chain Reaction , Recombinant Fusion Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/isolation & purification , Solubility , Substrate Specificity
20.
Cancer Res ; 62(6): 1797-801, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11912157

ABSTRACT

Resistance to chemotherapy targeting microtubules could be partially because of the delay in chromosome condensation and segregation during mitosis. The Chfr pathway has been defined recently, and its activation causes a delay in chromosome condensation in response to mitotic stress. Because Chfr contains a RING-finger domain, we tested whether Chfr inhibits chromosome condensation through an ubiquitin (ubiquitin)-dependent pathway. In the presence of purified E1, Ubc4, or Ubc5, and ubiquitin, Chfr catalyzes its own ubiquitination in vitro, an activity requiring the RING domain. In vivo, overexpressed Chfr but not a RING domain mutant is spontaneously ubiquitinated. Our studies with DLD1 cells stably expressing wild-type Chfr and Chfr lacking the RING domain indicated that the RING-finger deletion mutant was defective in inhibiting chromosome condensation after Taxol treatment. In addition, Chfr expression increases the survival rate after Taxol treatment, an activity requiring the RING domain. Preliminary studies indicate that Chfr expression is cell cycle regulated and is dependent on its ubiquitin ligase activity. It is very likely that the Chfr-mediated ubiquitin-dependent pathway is a critical component of the response to mitotic stress.


Subject(s)
Cell Cycle Proteins/physiology , Ligases/metabolism , Mitosis/physiology , Neoplasm Proteins , Ubiquitin/metabolism , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , DNA Damage , Humans , Mitosis/drug effects , Molecular Sequence Data , Paclitaxel/pharmacology , Poly-ADP-Ribose Binding Proteins , Protein Structure, Tertiary , Stress, Physiological , Topotecan/pharmacology , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...