Direct cholesterol esterase assay

A direct assay for cholesterol esterase is provided wherein the assay reagent comprises a tetrazolium salt, a cholesterol ester an exogenous electron carrier to create an assay sample. In one embodiment the reagent is mixed with a test sample and the presence of cholesterol esterase is detected by an optical response. In a second embodiment, the reagent is mixed with a test sample and the optical response is quantitated by comparison with standards to determine the cholesterol esterase activity in the test sample. Kits are also provided which comprise the reagent components.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
FIELD OF INVENTION

[0001] The present invention relates to a method of detection of enzymatic chemical reactions that result in the cleavage or formation of a chemical usually covalent bond. More particularly, the invention is a method for the chromogenic or fluorogenic detection of such enzyme reactions, in particular as an assay screen for new chemical combinations that are produced by biotechnology methodologies, that may have activity in enzyme-substrate interactions.

[0002] 1. Background of the Invention

[0003] Enzymes are catalytic proteins that are pervasive in biological systems. Many enzymes catalyze specific reactions which entail the cleavage or formation of a chemical bonds In particular such an Enzyme (E) will increase the rate of reaction of a specific Substrate (S) that involves the formation or cleavage of a covalent bond resulting in a Product (P). Enzymes are necessary in almost every biological reaction, and helpful in many chemical, pharmaceutical and manufacturing processes. Detecting enzyme activity and defining and measuring enzyme-substrate interactions is desirable in many clinical and laboratory situations, particularly in screening enzyme activity and screening molecules as inhibitors, enhancers or modifiers of pharmacologically interesting enzymes.

[0004] 2. Description of Related Art

[0005] Known enzymes are classified by the International Union of Biochemistry Commission on Enzymes into six distinct categories: oxdoreductases, transferases, hydrolases, lyases, isomerases and ligases. Recent advances in enzymology have identified previously unknown and/or non-natural catalytic molecules that have enzymatic-like speed and specificity, such as extremozymes, abzymes, recombinant enzymes, semi-synthetic enzymes, and catalytic ribozymes.

[0006] Recently techniques have been developed which permit large numbers of different chemical compounds to be synthesized rapidly and systematically for drug screening. Large collections of such compounds called, combinatorial chemical libraries, are expensive to produce so that typically only milligram quantities or less of each different molecule is in a library. Screens for different types of pharmacological or chemical activity can generally require different techniques, different instruments, varying time frames, different sensitivity levels, different software and different methods of data interpretation. As a result, to screen a large combinatorial library, or other large colon of compounds for different types of pharmaceutical or chemical activity heretofore required great expenses for training, instrumentation and reagents. Sifting through such libraries of molecules to determine structural features which show activity and act as possible pharmacological or industrial agents is a tremendous effort.

[0007] The field of enzyme study dates back more than one hundred years. Many methods to study and detect enzymatic events are now known. Significantly important enzyme-assay methods include: (1) spectrophotometry, using either ultraviolet or visual light; (2) fluorometry; (3) assays involving detection of radioactivity; (4) coupled assays; and (5) enzyme linked immunsorbent assays (ELISA). Virtually every enzyme requires a specific and unique substrate for its reactivity. The development of an assay for a particular enzyme/substrate reaction is often a difficult endeavor. Because this is a mature field—although still the subject of intense research and development activities—many textbooks and compilations of methods exist, in addition to articles in peer-reviewed journals.

[0008] Conventional assays for enzyme activity are virtually as numerous as the number of enzymes. Some examples of conventional assays for enzyme activity are:

[0009] creatine kinase, which is used as a serum control for the diagnosis of muscle deterioration is most frequently assayed in a coupled system with pyruvate kinase and lactate dehydrogenase;

[0010] proteases are conventionally measured using specific synthetic substrates which contain a chromogenic or fluorogenic enzyme conjugate at the amide bond which is hydrolyzed by the enzyme;

[0011] chloramphenicol acetyl transferase (CAT) is a widely used reporter gene in expression studies. There are several commercially available assays. Such assays for CAT include enzyme linked assays (ELISA), radioactive assays and fluorescent methods. Conventional ELISA methods for assaying for CAT typically take from 2-4 hours and are generally sensitive to only 1-2.5×10−12 g/ml of enzyme. The radioactive and fluorescent assays use expensive and/or dangerous reagents, and typically require a time-consuming post-event separation to measure the CAT activity.

[0012] Due to ease of use, specificity and sensitivity, the current method of choice for detection in most assay systems is radioactivity. However the rapid decay of the radioactive probe, danger of radiation exposure, extensive processing of samples, and storage and disposal problems for radioactive materials make non-radioactive methods of detection desirable.

[0013] For conventional non-radioactive detection systems to work, synthetic substrates must be designed to report on the event being monitored. In the case of many proteases, hydrolysis products serve to report on the activity of the protease. However, such hydrolysis products are frequently carcinogenic.

[0014] The design of non-radioactive methods usually involves the attachment of chromophores, fluorophores or lumigens at the scissile bond of the reporter substrate. A signal is generated if the enzymatic event takes place because a detectable chromogenic, fluorogenic or lumigenic species is liberated. Designing such reporter substrates is difficult; when a probe is introduced onto the substrate, the substrate can lose its lock and key fit to the enzyme, thus losing its enzyme specificity. If the substrate still fits the enzyme, the binding and energetics of the enzyme catalysis may be altered in significant ways with the result that the synthetic reporter substrate will not be a true measure of the enzyme reaction.

[0015] Proteases are enzymes that hydrolyze proteins. All living organisms contain proteases to metabolize proteins, regulate cellular processes, defend themselves against exogenous proteins and mediate other important requirements of survival. In the purification of any protein from a natural source, it is necessary to inhibit endogenous proteases to prevent them from breaking down the proteins of interest. The search for the presence of known and unknown proteases in a sample is an important endeavor both at the research stage and the development stage in all areas of biotechnology and related sciences. If the protease is known and it is known to be in the sample of interest, methods for its analysis and strategies for its purification or inhibition will generally have been elaborated. If a sample contains a protease which is not known or, which is not known to be present in that sample, it may be difficult to determine that one is even present in that sample and considerably more difficult to determine what type of protease it is in order to inhibit its activity.

[0016] Proteases are classified into a number of categories dependent upon their mechanism of action. They are further classified dependant upon the cleavage site in the proteins which they hydrolyze. The combination of action and cleavage site leads to multiplicative complexity in determining what proteases are present and how their activities might be inhibited. There is no single cleavage site in a protein which will be hydrolyzed by all enzymes and therefore no known substrate which will be a reporter for all enzymes. More importantly, there is no rapid, sensitive high-throughput method to characterize many types of enzyme activity in a single experiment.

[0017] Existing art is aimed at determining if any protease activity is present in a sample and is limited to two types of tests:

[0018] a. Fluorescently labeled casein;

[0019] b. Electrophoretic mobility assay.

[0020] Each of these assays typically requires lengthy incubation, is therefore slow (the test may take hours). Moreover the assays are generally insensitive, and may require post-hydrolysis separation or expensive instrumentation. In the best of circumstances and either assay will tell if protease activity is present, but cannot tell what type of protease is present, whether there is more than one type of protease present, or how to analyze or inhibit the protease based on known enzyme-substrate interaction.

[0021] Tetrazolium salts have been used if the study of the mitochondrial respiratory chain in vivo. A reduction of a substrate by an enzyme produces electrons which are transferred to the tetrazolium salt yielding a formazan which is deeply colored. The tetrazolium salt thus functions as an indication. The use of an exogenous electron carrier such as phenazine methosulfate can significantly increase the speed and sensitivity of the reaction. U.S. Pat. No. 5,354,658 entitled “Non-Radioactive Method for a Labelled Segment and A Solution or Composition Therefor”, the disclosure of which is hereby incorporated by reference, describes a sensitive and specific method of using phenazine methosulfate and dimethylthiazol diphenyl tetrazolium (MTT) to detect a specific enzyme-substrate in particular the reaction of alkaline phosphatase with a 5-bromo-4-chloro-3-indolyl phosphate reaction in vitro. The method of the patent allows a thousand times greater increase of sensitivity, and orders of magnitude greater speed than previously reported tetrazolium methods. However this patent did not address or solve the problem of screening large combinatorial chemical libraries rapidly and efficiently, much less teach or suggest any solution to that problem.

SUMMARY OF THE INVENTION

[0022] According to the present invention there is provided a non-radioactive method of monitoring an enzyme-substrate reaction by addition of an exogenous electron carrier and a tetrazolium salt to the reaction medium, and allowing the reaction to proceed to a colored or fluorescent formazan in an irreversible reaction. Preferred tetrazolium salts for the invention have the structure of (1) where R is cyano, aryl, heteroaryl or aralkyl; R1 is aryl, heteroaryl, or aralkyl and R2 is aryl, heteroaryl or aralkyl including: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MMT); 2,2′-di-p-nitrophenyl-5,5′-diphenyl-3,3′-(3,3′-dimethoxy-4,4′-diphenylene)ditetrazolium chloride (nitroblue tetrazolium, NBT); 2,3,5-triphenyl tetrazolium chloride (TTC); 2-(2′-benzothiazolyl)-5-styryl-3-(4phthalhydrazidyl) tetrazolium chloride (BPST); neotetrazolium chloride (NTC); 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2-H-tetrazolium-5-carboxanilide inner salt (XTT); p-iodonitrotetrazolium violet (INT) and cyanoditolyl tetrazolium chloride (CTC). Dimers of tetrazolium salts also are useful to this invention. It is anticipated that dimethylthiazol diphenyl tetrazolium (MTT) as the tetrazolium salt will be particularly preferred in this invention.

[0023] Preferred exogenous electron carriers for the invention include: phenazine methosulfate (PMS); phenazine ethosulfate (PES), nicotinamide adenine dinucleotide (NAD); nicotinamide adenine dinucleotide; flavin adenine dinuecleotide (FAD) or 4-aminoantipyrine.

[0024] The tetrazole-catalyst color indicator test can be linked to a wide variety of different enzyme-substrate electron transfers.

[0025] The present invention also provides for a solution or composition to be used as a test kit. The test kit of the invention would include a substrate, a tetrazolium salt and an electron carrier which when in solution added to a sample to be assayed would be capable of producing a colored or fluorescent formazan which results in a color or fluorescent change indicate of an electron transfer. Alternatively, the test kit could include one or more substrates, one or more enzymes, one or more cofactors a tetrazolium salt, an electron carrier and one or more specific inhibitors which when in solution added to a sample to be assayed would be capable of producing a colored or fluorescent formazan which results in a color or fluorescent change indicative of an electron transfer.

[0026] Preferred embodiments of the invention provide for a chromogenic or fluorogenic method that is non-radioactive and can be amenable to existing instrumentation, and software packages for enzyme analysis. The preferred methods of the invention have considerably high sensitivity, namely a sensitivity of 10−15 gram or less in contrast to prior methods of 10−12 i.e. 103 or 1,000 times more sensitive. Moreover, preferred embodiments of the invention can achieve a detectable color change in five to fifteen minutes. No amplification technique of the formazan product is required in preferred embodiments of the invention. No stabilizing agent for the tetrazolium salt is required in preferred embodiments.

[0027] The present invention permits different enzyme to be studied using the same spectrophotometer or plate reader, the same robotic system, the same training or personnel, similar or identical reagent systems, similar or identical disposables and consumables, similar or identical software, and similar or identical calculations of activity. Preferred embodiments of the present invention may be used to particular advantage in carrying out screening for useful chemical activities of the compounds of chemical libraries since all screens for various activities of interest using the tetrazolium and electron carrier will deliver the identical detection molecule, with the same wavelength of detection, the same extinction coefficient, similar reaction times and similar sensitivity and detection levels. In addition, with proper planning, the use of inhibitors or other chemicals, will allow the possibility of two or more entirely different screens to be done in the same microwell or curvette or on the same blot.

[0028] The invention described herein has many advantages over the previous methods of detection, including the use of natural substrates, the ability to assay a wide variety of enzymes, presumably even presently unknown enzymes, to detect activity, and the ability to tailor the reaction to a specific determination of one enzyme in the presence of competing enzymatic activities.

DESCRIPTION OF THE PREFERRED EMBODIMENTS

[0029] In one preferred embodiment of the invention, enzyme-substrate reactions are detected by transfer of an electron to dimethylthiazol diphenyl tetrazolium bromide. Dimethylthiazol diphenyl tetrazolium bromide has the chemical name of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2-H-tetrazolium bromide, the chemical formula, C18H16N5SBr, a molecular weight of 414.33, a melting point of 195° (dec), and an absorption at &lgr;MAX 378 nm.

[0030] The preferred exogenous electron carrier phenazine methosulfate has the chemical formula of N-methylphenazonium methosulfate, the chemical formula C14N14N2O4S, a molecular weight of 306.34 and a melting point between 158°-160° (dec), and an absorption at &lgr;max 386 nm.

[0031] In a preferred assay of the invention, the enzyme chloramphenicol acetyl transferase (CAT enzyme) reacts with chloramphenicol and aryl CoA in the presence of the indicator dimethylthiazol tetrazolium (MTT) and phenazine methosulfate (PMS). The MTT serves as a hydrogen acceptor in the reaction. The reaction can be written as:

CAT+chloramphenicol+acyl coA→CAT-chloramphenicol-CoA CAT-chloramphenicol-CoA+PMS→CAT+PMS.H2+acyl-chloramphenicol+HS-CoA PMS-H2+MTT→formazan+PMS.

[0032] The formazan product is colored and may therefore be detected in the presence of the other reaction species, which are generally colorless.

[0033] While an assay for chloramphenicol acetyl transferase, as desired above, is a preferred application of the invention the method may be used to detect substantially all categories of enzyme activity, for example, oxdoreductases, transferases, hydrolases, lyases, isomerases, ligases and novel classes of enzymes. Consequently, the enzyme activity assay of the present invention is expected to have applicability in both commercial processes such as pharmaceutical development, insect control, food science, pulp and paper, laundry and other industrial processes and basic molecular scientific research.

[0034] In addition, with this invention, test kits for specific enzyme/substrate interactions could be provided that include specific substrates and a tetrazolium dye. Individually, the different substrates would have different spectral characteristics and therefore could not be detected conventionally by the same spectrophotometer or microplate reader at the same time, same wavelength and same extinction coefficient. In contrast, with the present invention, different substrates ultimately yield the same reduced formazan, so that multiple enzymes can be studied with the same spectroscopic settings. A single microplate could have wells for each specific substrate. Combinations of inhibitors and control enzymes could be used to define new or unexpected enzymatic activities. Observing the development of color would confirm the presence or absence of specific enzymes.

[0035] The unique feature of this invention is that widely different enzymes, widely different inhibitors and widely different specific substrates can be studied with the same tetrazolium detection reagent and the same spectrophotometer or plate reader. This adds a tremendous efficiency over current art.

EXAMPLES

[0036] The generalized reaction where E=enzyme, S=substrate, T=tetrazolium salt, C=electron carrier, P=product is:

E+S→E−S

E−S+T→E+TH2+P

[0037] or

E+S→E−S

E−S+C→E+CH2+P

CH2+T→C+TH2

[0038] The experimental results for Experiments 1-12 are described by the color change of the experimental solution. These colors result from the interaction of the enzyme and substrate with the transfer of an electron to a tetrazolium salt. The reactive product, formazan, is detected as dark blue, black. The BCI substrate without MTT is pale blue. PMS and MTT alone are yellow. PMS and BCI is yellow+blue which is green. The reaction detected required the enzyme, substrate and tetrazolium salt.

Example 1

[0039] Microtiter testing was performed, with the following substrate combinations to demonstrate chromogenic detection of esterase (EC3.1.1.1). 1 Reagents PBS Phosphate Buffer Solution MTT Dimethylthiazol Tetrazolium (Sigma M-2128) PMS Phenazine methosulfate (Sigma P-9625) BCI acetate 5-Bromo-4-chloro-3-indolyl acetate (Sigma B-4377) in 50% Dimethyl formamide Esterase diluted with PBS @ pH 7.4 to a final concentration of 100 units/ml (Sigma E-2884)

[0040] Experiment:

[0041] Four test samples were prepared. The samples A, B, C, D contained the following reactants: 2 A. 100 &mgr;l PBS (Phosphate Buffer Solution) @ pH 7.4 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI acetate @ 12.5 mg/ml B. 100 &mgr;l PBS @ pH 7.4 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI acetate @ 12.5 mg/ml C. 100 &mgr;l PBS @ pH 7.4 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI acetate @ 12.5 mg/ml D. 100 &mgr;l PBS @ pH 7.4 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI acetate @ 12.5 mg/ml

[0042] 10 &mgr;l of H2O was added to substrate sample A to serve as a control 10 &mgr;l of the enzyme esterase (1 unit) was added to substrate samples B, C, and D. Detection of test results was done by visual determination. 3 Results Sample Contents Reaction A MTT, PMS, BCI acetate yellow - no color change (no esterase) detected B MTT, H2O, BCI acetate, 10 &mgr;l dark blue < 10 seconds) esterase C H2O, PMS, BCI acetate, 10 &mgr;l yellow-blue - slight change esterase D MTT, PMS, BCI acetate, dark blue (instantaneous) 10 &mgr;l esterase

Example 2

[0043] Microtitre testing was performed, with the following substrate combinations—in duplicate— to demonstrate chromogenic detection of esterase (EC 3.1.1.1): 4 Reagents: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-2128) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) BCI acetate 5-Bromo-4-chloro-3-indolyl-acetate (Sigma B 4377) in 1 ml of H2O to a 5 mg/ml concentration BCI butyrate 5-Bromo-4-chloro-3-indolyl-butyrate (Sigma B 9151) in 1 ml of H2O to a 5 mg/ml concentration Esterase Diluted with H2O to a 5 mg/ml concentration (Sigma E-2884) Experiment: A. 100 &mgr;l de-ionized H2O A′. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate B. 100 &mgr;l de-ionized H2O B′. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l H2O 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate C. 100 &mgr;l de-ionized H2O C′. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate D. 100 &mgr;l de-ionized H2O D′. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate E. 120 &mgr;l de-ionized H2O E′. 120 &mgr;l de-ionized H2O 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate

[0044] 10 &mgr;l of H2O was added to substrate sample A and A′ to serve as a control. 10 &mgr;l of esterase (1 unit) was added to substrate samples B, B′, C, C′, D, D′, and E, E′. 5 Results Sample Contents Reaction A MTT, PMS, BCI-butyrate yellow - no color change (no esterase) detected B MTT, H2O, BCI-butyrate dark blue (<30 minutes) (esterase) C H2O, PMS, BCI-butyrate yellow-blue (<30 minutes) (esterase) D MTT, PMS, BCI-butyrate dark blue (<20 minutes) (esterase) E BCI-butyrate (esterase) pale blue (>1 hour) A′ MTT, PMS, BCI-acetate yellow - no color change (no esterase) detected B′ MTT, H2O, BCI-acetate dark blue (<10 sec) (esterase) C′ H2O, PMS, BCI-acetate yellow-blue (<30 sec) (esterase) D′ MTT, PMS, BCI-acetate dark blue (instant detection) (esterase) E′ BCI-acetate (esterase) pale blue (>10 minutes)

[0045] All chromogenic BCI-butyrate reactions were observed to be much slower than the corresponding BCI-acetate reactions, event though the concentration of substrates and enzymes were similar. This indicates that the acetate reactions are a better substrate for esterase. Time to react could be used to qualify different substrates in unknown samples.

Example 3

[0046] Microtiter testing was performed, with the following substrate combinations to demonstrate chromogenic detection of esterase (EC3.1.1.). 6 Reagents MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-2128) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) BCI acetate 5-Bromo-4-chloro-3-indolyl 1,3 diacetate (Sigma B-5630) in H2O (5 mg/ml) Esterase diluted with H2O to a final concentration of 100 units/ml (Sigma E-2884)

[0047] Experiment:

[0048] Five test samples were prepared. The samples A, B, C, D, E contained the following reactants: 7 A. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-1,3 diacetate B. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI-1,3 diacetate C. 100 &mgr;l de-ionized H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI-1,3 diacetate D. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-1,3 diacetate E. 120 &mgr;l de-ionized H2O 10 &mgr;l BCI-1,3 diacetate

[0049] 10 &mgr;l of H2O was added to substrate sample A to serve as a control. 10 &mgr;l of the enzyme esterase (1 unit) was added to substrate samples B, C, D and E. Detection of test results were done by visual determination. 8 Results Sample Contents Reaction A MTT, PMS, BCI-1,2 diacetate yellow - no color change (no esterase) detected B MTT, H2O, BCI-1,3 dark blue (>1 hour) diacetate, 10 &mgr;l esterase C H2O, PMS, BCI-1,3 diacetate, yellow-blue 10 &mgr;l esterase D MTT, PMS, BCI-1,3 dark blue (>40 minutes) diacetate, 10 &mgr;l esterase E H2O, BCI-1,3 diacetate pale faint blue

[0050] All chromogenic BCI-1,3 diacetate reactions were observed to be much slower than the corresponding BCI-acetate and slower than the BCI-butyrate reactions. Of the BCI-substrates tested, substrate, preference for both esterase and cholesterol esterase is BCI-acetate>BCI-butyrate> BCI-1,3 diacetate, the concentration of substrates and enzymes were similar. Using a series of substrates which are good, better and best for a set of enzymes, it is possible to distinguish relative activity or presence of one or more enzymes.

Example 4

[0051] Microtitre testing was performed, with the following substrate combinations to demonstrate chromogenic detection of B-glucuronidase (EC 3.2.1.31). 9 Reagents PBS Phosphate Buffer Solution as aqueous solvent and to maintain pH in the 5.0 range MTT Dimethylthiazol Tetrazolium (Sigma M-2128) PMS Phenazine methosulfate (Sigma P-9625) BCI 5-Bromo-4-chloro-3-indolyl-B-D-glucuronide glucuronide (Sigma B-5285) Reconstituted with 1 ml H2O to a final concentration of 10 mg/ml Beta Reconstituted with 1 ml H2O to a final concentration glucuronidase of 0.1 unit/&mgr;l (Sigma G-5897)

[0052] Experiment:

[0053] Four test samples were prepared. The samples A, B, C, D contained the following reactants: 10 A. 100 &mgr;l PBS (Phosphate Buffer Solution) @ pH 5.0 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI glucuronide B. 100 &mgr;l PBS @ pH 5.0 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI glucuronide C. 100 &mgr;l PBS @ pH 5.0 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI glucuronide D. 100 &mgr;l PBS @ pH 5.0 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI glucuronide E. 100 &mgr;l PBS @ pH 5.0 10 &mgr;l H2O 10 &mgr;l BCI glucuronide

[0054] 10 &mgr;l of H2O was added to substrate sample A to serve as a control. 10 &mgr;l of &bgr;-glucuronidase (10 units) was added to substrate samples B, C, and D. 11 Results: Sample Contents Reaction A MTT, PMS, BCI glucuronide yellow - no color change (no glucuronidase) detected B MTT, H2O, BCI glucuronide dark blue (<5 minutes) (glucuronidase) C H2O, PMS, BCI glucuronide yellow-blue - slight change (glucuronidase) D MTT, PMS, BCI glucuronide dark blue (<5 minutes) (glucuronidase) E H2O, BCI glucuronide pale blue (after 5 minutes) (glucuronidase)

Example 5

[0055] Microtitre testing was performed with a protease inhibitor and various substrate combinations to demonstrate esterase activity and/or contamination in a commercially available elastase (EC 3.4.21.36) preparation.

[0056] The esterase substrates were observed to generate positive signal upon addition of an elastase dilution (10 &mgr;l). Elastase was serially diluted twofold to signal extinction with elastase substrate. The last dilution yielding robust signal was incubated with the competitive inhibitor elastatinal for 10 minutes—with subsequent addition of esterase substrates. 12 Reagents: PBS Phosphate Buffer Solution MTT Dimethylthiazol Tetrazolium (Sigma M-2128) PMS Phenazine methosulfate (Sigma P-9625) BCI acetate 5-Bromo-4-chloro-3-indolyl acetate (Sigma B-4377) in 50% Dimethy formamide Elastase (Sigma lot # 17H8005) Elastase N-succinyl-ALA-ALA-ALA p nitroanilide in 50% Dimethy Substrate formamide (Sigma-4760) 50% DMF at a 12.5 mg/ml concentration Elastatinal Elastase inhibitor (Sigma E-0881

[0057] 13 Results: Enzyme + Contents Reaction Elastase + MTT, PMS, BCI-acetate dark blue (<5 minutes) Inhibitor Elastase + Elastase substrate clear (no color >10 min.) Inhibitor Elastase + MTT, PMS, BCI-acetate dark blue (<5 minutes) H2O Elastase + Elastase substrate yellow H2O

[0058] The generation of positive signal with esterase substrates and generation of positive signal in the microtitre wells containing elastase with elastase substrate and no initial signal development in wells containing the elastase—inhibitor reacted with elastase substrate demonstrate the presence of esterase activity and/or contamination in the elastase preparation.

Example 6

[0059] Microtitre testing was performed, with the following substrate combinations—in duplicate— to demonstrate chromogenic detection of beta-glucosidase (EC 3.2.1.21): 14 Reagents: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-2128) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) BCI glucoside 5-Bromo-4-chloro-3-indolyl-B-D-glucoside (Sigma M-4527) in 1 ml of H2O to a final concentration of 5 mg/ml Beta glucosidase Diluted with 10 ml of H2O to a final concentration of 50 units/ml (Sigma lot #37H4031)

[0060] Five test samples were prepared. The samples A, B, C, D, E contained the following reactants: 15 Experiment: A. 100 &mgr;l deionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI glucoside @ 5 mg/ml B. 100 &mgr;l deionized H2O 10 &mgr;l MTT(10 mM) 10 &mgr;l H2O 10 &mgr;l BCI glucoside @ 5 mg/ml C. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI glucoside @ 5 mg/ml D. 100 &mgr;l deionized H2O 10 &mgr;l MTT(10 mM) 10 &mgr;l PMS(10 mM) 10 &mgr;l BCI glucoside @ 5 mg/ml E. 120 &mgr;l de-ionized H2O 10 &mgr;l BCI glucoside @ 5 mg/ml

[0061] 10 &mgr;l of H2O was added to substrate sample A to serve as a control. 10 &mgr;l of &bgr;-glucosidase (0.5 units) was added to substrate samples B, C, D and E. 16 Results: Sample Contents Reaction A MTT, PMS, BCI-glucoside yellow - no color (no glucosidase) change detected B MTT, H2O, BCI-glucoside dark blue (<5 minutes) (glucosidase) C H2O, PMS, BCI-glucoside yellow-blue - slight change (glucosidase) D MTT, PMS, BCI-glucoside dark blue (<5 minutes) (glucosidase) E BCI-glucoside (glucosidase) pale blue (>10 minutes)

[0062] The presence of enzyme and substrate was rapidly detected, with or without the electron transport carrier PMS.

[0063] Blot testing on 0.2 um nitrocellulose membrane was performed to demonstrate chromogenic detection of beta glucosidase. 5 ul of glucosidase (0.25U) was spotted and allowed to dry. 10 ul of each substrate combination (B, C, D and E) was applied to the dried enzyme spots. Similar detection results were obtained as above. Controls were tested with 5 ul H2O spots—instead of enzyme—with respective substrate combinations (10 ul)—with no observable detection reaction.

Example 7

[0064] Microtiter testing was performed, with identical protocol and substrate combinations of example 5 (in duplicate) to demonstrate chromogenic detection of cholesterol esterase (EC3.1.1.13). 17 Reagents: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-2128) PMS (10 mM) Phenazine methosulfate (Sigma P-9635) BCI 5-Bromo-4-chloro-3-indolyl-B-D-acetate (Sigma B 4377) acetate in 1 ml of H2O BCI butyrate 5-Bromo-4-chloro-3-indolyl-B-D-butyrate (Sigma B 9151) in 1 ml of H2O to a 5 mg/ml concentration Cholesterol reconstituted to a concentration of esterase 5 U/ml with H2O (Sigma C-5921)

[0065] Experiment:

[0066] 10 &mgr;l of H2O was added to substrate sample A to serve as a control. 10 ul of cholesterol oxidase (25 U/ml.) was added in appropriate testing. 18 A. 100 &mgr;l de-ionized H2O A'. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate B. 100 &mgr;l de-ionized H2O B'. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l H2O 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate C. 100 &mgr;l deionized H2O C'. 100 &mgr;l deionized H2O 10 &mgr;l H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate D. 100 &mgr;l de-ionized H2O D'. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS(10 mM) 10 &mgr;l PMS(10 mM) 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate E. 120 &mgr;l de-ionized H2O E'. 120 &mgr;l de-ionized H2O 10 &mgr;l BCI butyrate 10 &mgr;l BCI acetate

[0067] 10 &mgr;l of H2O was added to substrate sample A and A′ to serve as controls. 10 ul of esterase (1 unit) was added to substrate samples B, B′, C, C′, D, D′ and E, E′. 19 Results: Sample Contents Reaction A MTT, PMS, BCI-butyrate yellow - no color change (no cholesterol esterase) detected B MTT, H2O, BCI-butyrate dark blue (<30 minutes) (cholesterol esterase (0.05 U)) C H2O, PMS, BCI-butyrate yellow-blue (<30 minutes) (cholesterol esterase (0.05 U)) D MTT, PMS, BCI-butyrate dark blue (<20 minutes) (cholesterol esterase (0.05 U)) E BCI-butyrate (cholesterol pale blue (>1 hour) esterase (0.05 U)) A' MTT, PMS, BCI-acetate yellow - no color change (no cholesterol esterase) detected B' MTT, H2O,BCI-acetate dark blue (<10 sec) (cholesterol esterase (0.05 U) C' H2O, PMS, BCI-acetate yellow-blue (<30 see) (cholesterol esterase (0.05 U) D' MTT, PMS, BCI-acetate dark blue (instant detection) (cholesterol esterase (0.05 U) E' BCI-acetate (cholesterol pale blue (>10 minutes) esterase (0.05 U)

[0068] All chromogenic BCI-butyrate reactions were observed to be much slower than the corresponding BCI-acetate reactions, event though the concentration of substrates and enzymes were similar. This indicates that the acetate reactions are a better substrate for esterase. Time to react could be used to qualify different substrates in unknown samples.

Example 8

[0069] Microtitre testing was performed, with the following substrate combinations—in duplicate— to demonstrate chromogenic detection of cholesterol oxidase (EC1.1.3.6): 20 Reagents: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-66H5033) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) cholesterol oxidase reconstitute with deionized H2O to a concentration of 25 U/ml (Sigma C-5421) Cholesterol Std reconstituted with deionized H2O to a concentration (Sigma C-9908) 50 mg/dl Experiment A. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l Cholesterol Std @ 50 mg/dl B. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l Cholesterol Std @ 50 mg/dl C. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l Cholesterol Std @ 50 mg/dl D. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l Cholesterol Std @ 50 mg/dl E. 120 &mgr;l de-ionized H2O 10 &mgr;l Cholesterol Std @ 50 mg/dl

[0070] 10 &mgr;l of H2O was added to substrate sample A (substrate control) Cholesterol Oxidase (25 U/ml) was added to A, B, C, D and E. 21 Results: Sample Contents Reaction A MTT, PMS, cholesterol yellow, then slight green-yellow (no Cholesterol Oxidase) B MTT, H2O, cholesterol yellow, no color change (Cholesterol Oxidase) C H2O, PMS, cholesterol yellow, no color change (Cholesterol Oxidase) D MTT, PMS, cholesterol dark blue(<1 minutes) (Cholesterol Oxidase) E H2O (Cholesterol Oxidase) clear, no color change

Example 9

[0071] Microtitre testing was performed with the following substrate combinations in duplicate to demonstrate chromogenic detection of glucose oxidase (EC 1.1.3.4): 22 Reagents: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-66H5033) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) glucose oxidase diluted with H2O to a concentration of 0.2 U/&mgr;l (Sigma G-9010)) glucose solubilized with H2O to a concentration (Sigma G-8270) 50 mg/dl Experiment: A. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l 5% glucose solution B. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l 5% glucose solution C. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l 5% glucose solution D. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l 5% glucose solution E. 120 &mgr;l de-ionized H2O 10 &mgr;l 5% glucose solution

[0072] 10 &mgr;l of H2O was added to substrate sample A as the control sample. Glucose oxidase (2U) was added to samples B,C,D. 23 Results: Sample Contents Reaction A MTT, PMS, glucose yellow, (no glucose oxidase) B MTT, H2O, glucose (glucose dark blue (>10 mm) oxidase) C H2O, PMS, glucose (glucose yellow-green oxidase) D MTT, PMS, glucose (glucose dark blue(immediate) oxidase)

Example 10

[0073] Microtitre testing was performed with the following substrate combinations in duplicate to demonstrate chromogenic detection of chloramphenicol acetyltransferase (EC 2.3.1.28) 24 Results: MTT (10mM) Dimethylthiazol Tetrazolium (Sigma M-66H5033) PMS (10mM) Phenazine methosulfate (Sigma P-9625) Chloramphenicol reconstituted with deionized H2O to a concentration of 500 U/ml Acetyltransferase (Sigma C-2900) Acetyl CoA solubilized with deionized H2O to a concentration of 2 mg/ml Experiment: A. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l Acetyl CoA 10 &mgr;l Chloramphenicol B. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT(10 mM) 10 &mgr;l H2O 10 &mgr;l Acetyl CoA 10 &mgr;l Chloramphenicol C. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l Acetyl CoA 10 &mgr;l Chloramphenicol D. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l Acetyl CoA 10 &mgr;l Chloramphenicol

[0074] 10 &mgr;l of H2O was added to substrate sample A as the substrate control. Chloramphenicol acetyl transferase (5U/10 &mgr;l) was added to each substrate combination. 25 Results: Sample Contents Reaction A MTT, PMS, yellow Chloramphenicol, Acetyl CoA (no Chloramphenicol acetyl transferase) B MTT, H2O, orange detection (>5 minutes) Chloramphenicol, Acetyl CoA (Chloramphenicol acetyl fransferase) C H2O, PMS, Chloramphenicol, orange - red (<2 minutes) Acetyl CoA (Chloramphenicol acetyl transferase) D MTT, PMS, Chloramphenicol, yellow Acetyl CoA (Chloramphenicol acetyl transferase)

[0075] Further testing was performed with ten-fold dilutions of the enzyme using substrate sample D. The reaction is sensitive to between 0.5U and 0.05U of enzyme.

Example 11

[0076] Microtitre testing was performed with the following substrate combinations in duplicate to demonstrate chromogenic detection of neuraminidase (EC 3.2.1.18) 26 Reagents: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-66H5033) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) BCI-acetylneuraminic 5-bromo-4-chloro-3-indolyl acetyl neuraminic acid acid diluted with 500 &mgr;l of H2O Neuraminidase Solubilized with 1 ml of H2O to a final concentra- tion of 10 units/ml Experiment: A. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-acetylneuraminic acid B. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI-acetylneuraminic acid C. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-acetylneuraminic acid D. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-acetylneuraminic acid E. 120 &mgr;l de-ionized H2O 10 &mgr;l BCI-acetylneuraminic acid

[0077] 10 &mgr;l of H2O was added to substrate sample A as the substrate control. 10 &mgr;l of neuraminidase (0.1 units) was added to substrate samples B, C, D and E. 27 Results: Sample Contents Reaction A MTT, PMS, BCI- yellow acetylneuraminic acid (no neuraminidase) B MTT, H2O BCI- light blue formazan reaction acetylneuraminic acid, neuraminidase C H2O, PMS, BCI- green acetylneuraminic acid, neuraminidase D MTT, PMS, BCI- dark blue (>5 minutes) acetylneuraminic acid (neuraminidase) E H2O BCI-acetylneuraminic pale blue (<2 minutes) acid (neuraminidase)

[0078] The presence of enzyme and substrate was rapidly detected the tetrazolium salt plus the electron transport carrier.

Example 12

[0079] Microtitre testing was performed, with the following substrate combinations—in duplicate— to demonstrate chromogenic detection of Beta-N-acetylglucosaminidase (EC 3.2.1.30): 28 Results: MTT (10 mM) Dimethylthiazol Tetrazolium (Sigma M-66H5033) PMS (10 mM) Phenazine methosulfate (Sigma P-9625) Beta-N- solubilized with H2O (12.5 U/ml) acetylglucoaminidase (Sigma A-2415) BCI-acetylglucosaminide solubilized with 2 ml H2O to a concentration 12.5 mg/ml (Sigma B-3041) Experiment: A. 100 &mgr;l deionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-acetylglucosaminide B. 100 &mgr;l de-ionized H2O 10 &mgr;l MTT (10 mM) 10 &mgr;l H2O 10 &mgr;l BCI-acetylglucosaminide C. 100 &mgr;l de-ionized H2O 10 &mgr;l H2O 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-acetylglucosaminide D. 100 &mgr;l deionized H2O 10 &mgr;l MtT (10 mM) 10 &mgr;l PMS (10 mM) 10 &mgr;l BCI-acetylglucosaminide E. 120 &mgr;l de-ionized H2O 10 &mgr;l BCI-acetylglucosaminide

[0080] 10 &mgr;l of H2O was added to substrate sample A (substrate control.) 10 &mgr;l Beta-N-acetylglucoaminidate (0.125 units) was added to A, B, C, D and E. 29 Results: Sample Contents Reaction A MTT, PMS, BCI- yellow acetylglucosaminide (no Beta-N- acetylglucoaminidase) B MTT, H2O, BCI- light blue formazan reaction acetylglucosaminide, Beta-N acetylglucoaminidase C H2O, PMS, BCI- green acetylglucosaminide, Beta-N acetylglucoaminidase D MTT, PMS, BCI- dark blue(<2 minutes) acetylglucosaminide (Beta-N acetylglucoaminidase) E BCI-acetylglucosaminide light blue (>5 minutes) (Beta-N-acetylglucoaminidase)

[0081] The presence of the enzyme and substrate was rapidly detected in the presence of PMS and MTT.

[0082] There are a wide variety of enzymes and inhibitors that can be used with this invention, The following are several examples of possible embodiments:

Example 13

[0083] Reagents:

[0084] X=substrate+MT+PMS

[0085] E=sample to be analyzed for enzyme activity

[0086] I1=TLCK-specific inhibitor for chymotrypsin

[0087] I2=TPCK-specific inhibitor for trypsin

[0088] I3=Elastinal-specific inhibitor of elastase

[0089] Experiments:

[0090] 1. E would be incubated with I1, then treated with X: a color should develop if protease other than trypsin present.

[0091] 2. E would be incubated with I2, then treated with X: a color should develop if protease other than chymotrypsin present.

[0092] 3. E would be incubated with I3, then treated with X: a color should develop if protease other than elastase present.

[0093] 4. E would be incubated with I1 and I2, then treated with X: a color if protease other than trypsin or chymotrypsin are present.

[0094] 5. E would be incubated with I1 and 3, then treated with X: a color should develop if protease other than trypsin or elastase are present.

[0095] 6. E would be incubated with I2 and I3, then treated with X: a color should develop if protease other than chymotrypsin or elastase are present.

[0096] In order to confirm these inhibition-based results, it is possible with this invention to make specific substrate solutions in which MTT and PMS are added to individual solutions of I1, I2 and I3 as defined above. These three different substrates have different spectral characteristics and cannot be read by the same spectrophotometer at the same time at the same wavelength and using the same extinction coefficient to calculate enzymatic activity. With this invention a single microplate could have wells for each inhibition study outlined above and each specific substrate as well. Observing the development of patterns of color would have confirmatory results on identification of the specific enzymes mentioned.

Example 14

[0097] For lipase enzyme detection on a blot assay, the blot could be impregnated with a solution of the enzyme. Allow to dry. Treat the blot with a solution of natural or synthetic glyceride or cholesterol ester, plus MTT, plus PMS. A color indicative of enzymatic activity should develop.

Example 15

[0098] For lipase enzyme detection in solution (microplate, tubes or cuvettes), add an enzyme solution or enzyme-antibody conjugate in solution to each microplate, tubes or cuvettes. Add a solution synthetic or natural glyceride or cholesteryl ester, plus MTT, plus PMS. A color indicative of enzymatic activity develops and could be read in a spectrophotometer or plate reader.

Example 16

[0099] For the study of lipase inhibition in solution, preincubate the enzyme solution with a solution of inhibitor. Add the substrate solution containing synthetic or natural glyceride or cholesterol ester, MTT and PMS. Observe the development of color kinetically in comparison to a blank solution which contains enzyme and substrate but no inhibitor.

Example 17

[0100] For aldolase enzyme detection on a blot assay, the blot could be impregnated with a solution of the enzyme. Allow to dry. Treat the blot with a solution of D-fructose-1,6-biphosphate, plus MTT, plus PMS. A color indicative of enzymatic activity develops.

Example 18

[0101] For aldolase enzyme detection in solution (microplate, tubes or cuvettes), add an enzyme solution or enzyme-antibody conjugate in solution to each microplate, tubes or cuvettes. Add a solution of D-fructose-1,6-biphosphate, plus MT, plus PMS. A color indicative of enzymatic activity develops and could be read in a spectrophotometer or plate reader.

Example 19

[0102] For the study of aldolase inhibition in solution, preincubate the enzyme solution with a solution of inhibitor. Add the substrate solution containing of D-fructose-1,6-biphosphate, MTT and PMS. Observe the development of color as it develops in comparison to a blank solution which contains enzyme and substrate but no inhibitor.

Example 20

[0103] For phosphoglucomutase detection on a blot assay, the blot could be impregnated with a solution of the enzyme. Allow to dry. Treat the blot with a solution of glucose-1-phosphate, plus MTT, plus PMS. A color indicative of enzymatic activity develops.

Example 21

[0104] For phosphoglucomutase enzyme detection in solution (microplate, tubes or cuvettes), add an enzyme solution or enzyme-antibody conjugate in solution to each microplate, tubes or cuvettes. Add a solution of glucose-1-phosphate, plus MTT, plus PMS. A color indicative of enzymatic activity develops and could be read in a spectrophotometer or plate reader.

Example 22

[0105] For the study of phosphoglucomutase inhibition in solution, preincubate the enzyme solution with a solution of inhibitor. Add the substrate solution containing glucose-1-phosphate, MTT and PMS. Observe the development of color as it develops in comparison to a blank solution which contains enzyme and substrate but no inhibitor.

Example 23

[0106] For DNA ligase enzyme detection on a blot assay, the blot could be impregnated with a solution of the enzyme. Allow to dry. Treat the blot with a solution of synthetic or natural DNA fragments, plus MTT, plus PMS. A color indicative of enzymatic activity develops.

Example 24

[0107] For DNA ligase enzyme detection in solution (microplate, tubes or cuvettes), add an enzyme solution or enzyme-antibody conjugate in solution to each microplate, tubes or cuvettes. Add a solution of synthetic or natural DNA fragments, plus MTT, plus PMS. A color indicative of enzymatic activity develops and could be read in a spectrophotometer or plate reader.

Example 25

[0108] For the study of DNA ligase inhibition in solution, preincubate the enzyme solution with a solution of inhibitor. Add the substrate solution containing synthetic or natural DNA fragments, MTT and PMS. Observe the development of color as it develops in comparison to a blank solution which contains enzyme and substrate but no inhibitor.

Example 26

[0109] For DNA ligase enzyme detection on a blot assay, the blot could be impregnated with a solution of the enzyme. Allow to dry. Treat the blot with a solution of synthetic or natural DNA fragments, plus MTT, plus PMS. A color indicative of enzymatic activity develops.

Example 26

[0110] For DNA ligase enzyme detection in solution (microplate, tubes or cuvettes), add an enzyme solution or enzyme-antibody conjugate in solution to each tube. Add a solution of synthetic or natural DNA fragments, plus MTT, plus PMS. A color indicative of enzymatic activity develops and could be read in a spectrophotometer or plate reader.

Example 27

[0111] For the study of DNA ligase inhibition in solution, preincubate the enzyme solution with a solution of inhibitor. Add the substrate solution containing synthetic or natural DNA fragments, MTT and PMS. Observe the development of color as it develops in comparison to a blank solution which contains enzyme and substrate but no inhibitor.

[0112] The invention has been described with respect to preferred embodiments. However, as those skilled in the art will recognize, modifications and variations in the specific embodiments which have been described and illustrated may be resorted to without departing from the spirit and scope of the invention as defined in the appended claims.

Claims

1. A non-radioactive method of detecting an enzyme-substrate reaction comprising coupling said reaction with a tetrazolium salt and the transfer of free electrons from the reaction to the tetrazolium salt to produce a single colored or fluorescent formazan wherein the improvement comprises assaying a plurality of compounds for enzyme activity.

2. The method of

claim 1 wherein said formazan produces a visual or instrumental monitoring at a single wavelength with a single extinction coefficient.

3. The method of

claim 1 wherein said tetrazolium salt has the structure of: 1
where R1 is aryl or heteroaryl, and R2 is aryl or heteroaryl, X can be any common counter ion.

4. The method according to

claim 1 wherein the tetrazolium salt is dimethylthiazol tetrazolium (MTT).

5. The method of

claim 1 wherein said test sample is further reacted with an exogenous electron carrier.

6. The method of

claim 6 wherein said exogenous electron carrier is a phenazine salt of the structure: 2
where R is an alkyl, Z is an alkyl, alkoxyl, halo, or nitro, and X is any common counter ion.

7. The compound of

claim 6 wherein the phenazine is Phenazine Methosulfate.

8. The method of

claim 1 where the enzyme is Esterase (EC3.1.1.1).

9. The method of

claim 1 wherein the enzyme is Beta-glucuronidase (EC 3.2.1.31).

10. The method of

claim 1 wherein the enzyme is Beta-glucosidase (EC 3.2.1.21).

11. The method of

claim 1 where the enzyme is Cholesterol esterase (EC 3.1.1.13).

12. The method of

claim 1 where the enzyme is Cholesterol oxidase (EC 1.1.3.6).

13. The method of

claim 1 where the enzyme is Glucose oxidase (EC 1.1.3.4).

14. The method of

claim 1 where the enzyme is Chloramphenicol acetyltransferase (EC 2.3.1.28).

15. The method of

claim 1 where the enzyme is Beta-N-acetylglucosaminidase (EC 3.2.1.30).

16. The method of

claim 1 where the enzyme is Neuraminidase (EC 3.2.1.18).

17. The method of

claim 1 wherein said reaction concerns two or more enzymes which can be detected with a single substrate, wherein that substrate contains one or more sites for enzymatic action.

18. The method of

claim 9 wherein said test sample is further reacted with an exogenous electron carrier.

19. The method of

claim 1 wherein said test sample contains two or more enzymes and two or more substrates.

20. The method of

claim 11 wherein said test sample is further reacted with an exogenous electron carrier.

21. The method of

claim 1 wherein said test sample contains two or more enzymes, two or more substrates, and one or more inhibitors of said enzymes.

22. The method of

claim 13 wherein said test sample is further reacted with an exogenous electron carrier.

23. A kit of materials for performing the method of detection of enzyme reaction, consisting of a vial or packet of tetrazolium salt in an amount sufficient, when reacted with said test sample, to produce a colored or fluorescent formazan or a color or fluorescent change indicative of the transfer of electrons in the sample.

24. A kit of materials for performing the method of detection of enzyme reaction, consisting of a vial or packet of tetrazolium salt and a vial or packet of exogenous electron carrier, in an amounts sufficient, when reacted with said test sample, to produce a colored or fluorescent formazan or a color or fluorescent change indicative of the transfer of electrons in the sample.

Patent History
Publication number: 20010014462
Type: Application
Filed: Dec 21, 2000
Publication Date: Aug 16, 2001
Inventors: Dennis Wright (Talahassee, FL), David J. Phelps (Stamford, CT)
Application Number: 09746527
Classifications
Current U.S. Class: Involving Esterase (435/19)
International Classification: C12Q001/37;