throbber
the amount of thrombin and the
`In addition to varying the time of incubation,
`temperature of incubation (up to 37°C) may also be varied to determine the optimum
`conditions for cleavage of a particularfitsion protein.
`
`ENZYMATIC CLEAVAGE OF MATRIX-BOUND GST FUSION PROTEINS
`
`In this alternate protocol, GST fusion proteins that contain a thrombin cleavage site are
`bound to glutathione-agarose as described in UNIT 16.7. Prior to elution feom the matrix,
`thrombin is added and the protein of interest is cleaved from the GST carrier. The cleaved
`protein is collected in the wash buffer and the GST carrier remains bound to the beads,
`permitting easy and efficient physical separation of the reaction products.
`
`ALTERNATE
`PROTOCOL 2
`
`Additional Materials
`
`For recipes, see Reagents and Solutions in this unit (or cross-referenced unit); for common stock
`solutions, see APPENDIX 2; for suppliers, see APPENDIX 4.
`
`GST fusion protein bound to glutathione-agarose beads (UNIT 16.7)
`1% (v/v) Triton X-100 in phosphate—buffered saline (PBS; APPENDIX 2)
`GST wash buffer: 50 mM Tris«Cl (pH 7.5)/150 mM NaCl
`GST elution buffer: 50 mM Tris-Cl (pH 8.0)/5 mM reduced glutathione
`20- or 50- ml screw-cap tube
`
`1. Wash GST fusion protein bound to glutathione-agarose beads with 20 vol of 1%
`Tréton X-100 in PBS, using a 20- or 50-ml screw-cap tube. Centrifuge
`sec in a
`tabletop centrifuge at 500 x g, room temperature, to pellet the beads. Carefully remove
`and discard the supernatant. Resuspend the beads in 20 vol Triton X-100 buffer and
`repeat wash.
`
`2. After the second centrifugation, carefully remove and discard the supernatant.
`Resuspend the beads in 20 vol GST wash buffer.
`
`3. Pellet the beads and discard the supernatant. Resuspend the beads in 20 vol thrombin
`cleavage buffer. Repeat the centrifugation and resuspend the beads in S1 ml thrombin
`cleavage buffer.
`
`Although it is easier to wash the beads in large volumes, the amount of thrombin
`cleavage buffer to use in the cleavage reaction is best kept to a minimum.
`
`4. Remove a small aliquot of resuspended beads and add an equal volume of 2x SDS
`sample buffer. Store at —20°C until analyzed by SDS-PAGE (step 7).
`
`This sample is used to estimate the amount offitsion protein bound to the beads.
`
`5. Add thrombin to the remaining bead slurry at a ratio of 1% (W/w) thrombin to the
`estimated amount of bound fasion protein. Incubate 1 hr at 25°C.
`
`As in solution cleavage, the amount ofthrombin. time, and temperature of incubation
`can be adjusted to optimize the cleavage efficiency.
`
`6. Elute the cleaved and released protein by washing the beads with 1 bed volume of
`GST wash buffer. Centrifuge as in step 1 to pellet beads and collect supernatant.
`Repeat five times, but keep each wash fraction separate. Remove 20—|.Ll aliquots from
`each wash fraction for SDS-PAGE.
`
`7. Elute bound GST by repeating step 6 with GST elution buffer instead of GST wash
`buffer. Remove 20~ul aliquots from each fraction and analyze by SDS-PAGE to
`determine extent of cleavage. Include the aliquot of beads from step 4 on this gel.
`
`Current Protocols in Molecular Biology
`
`Protein
`Expression
`
`16.4.9
`
`Supplement 28
`
`BEQ 1016
`Page 190
`
`

`
`If cleavage is incomplete, the time of incubation and/or the amount of enzyme can be
`increased.
`
`Because this is an analytical-scale experiment, it is easiest to discard these beads after
`one use. Upon scaling up the procedure, the beads can be regenerated, as described
`in Reagents and Solutions, UNIT 16. 7.
`
`ALTERNATE
`PROTOCOL 3
`
`ENZYMATIC CLEAVAGE OF FUSION PROTEINS WITI-E
`ENTEROKINASE
`
`Enterokinase (also called enteropeptidase) is a mammalian trypsin-like serine protease
`that displays a high degree of specificity for the sequence (Asp)4-Lys, cleaving on the
`carboxy-terminal side of the lysine residue of the recognition sequence. Although in
`mammals the enzyme has evolved to recognize and cleave this sequence from the
`amino-termini of trypsinogens, it has been shown that enterokinase is also capable of
`cleaving fusion proteins that are expressed in bacteria and that contain this recognition
`sequence inserted between the carrier protein and the carboxy-terminal fusion partner.
`Enterokinase is capable of cleaving fusion proteins under a wide range of reaction
`conditions, with pH ranging from 4.5 to 9.5 and temperatures ranging from 4° to 45°C.
`Enterokinase is also extremely tolerant of the nature of the amino acid residue in the P1’
`position (except that the peptide bond between Lys-Pro at this position is totally refractory
`to cleavage; E. LaVallie and L. liacie, unpub. observ.). At sufficiently low ionic strength,
`enterokinase can cleave fusion proteins at a weight ratio of 1 2500 to 122000. At these ratios,
`typical cleavage reactions are carried out for 16 to 24 hr at 37°C, but these parameters
`(time, temperature, and enzyme/substrate ratio) can be adjusted as needed.
`
`The thioredoxin fusion vector pTRXFUS (UNI? 16.8) encodes an enterokinase cleavage site
`immediately preceding the polylinker cloning region. Proteins produced as Trx fusions
`using this system can be subsequently released by incubation with enterokinase, leaving
`their authentic amino-terminal sequence. The protocol below describes the use of bovine
`enterokinase in this application.
`
`Additional Materials
`
`For recipes, see Reagents and Solutions in this unit (or cross-referenced unit); for common stock
`solutions, see APPENDIX 2; for suppliers, see APPENDIX 4.
`
`1 mg/ml thioredoxin fusion protein (UNIT16.8) in 50 mM Tris-Cl (pH 8.0)/1 mM
`CaCl2
`10 ug/ml bovine enterokinase (Biozyme EK-3 grade) in 50 mM Tris-Cl
`(pH 8.0)/1 mM CaCl2
`
`NOTE: Many commercial preparations of enterokinase (bovine or porcine), with the
`exception of the source listed, are extremely impure and tend to be contaminated with,
`among other things, trypsin and chymotrypsin which can extensively degrade the fusion
`protein. It is recommended that only commercial enterokinase of the highest quality be
`used.
`
`1. Perform a pilot experiment to monitor the efficiency of cleavage with various ratios
`of enterokinase to fusion protein. Prepare five reactions:
`
`Reactions I to 4: 20 [11 of 1 mg/ml fusion protein, 1 [L1, 2 [L1, 5 it], and 10 11.1
`of 10 ug/ml bovine enterokinase, and 50 mM Tris-Cl (pH 8.0)/1 mM
`CaCl2, to a total of 30 ul.
`Reaction 5: 20 ul of 1 mg/ml fusion protein and 10 ul of 50 mM Tris-Cl
`(pH 8.0)/1 mM CaCl2 (mock digestion).
`
`Incubate samples 2 16 hr at 37°C.
`
`Current Protocols in Molecular Biology
`BEQ 1016
`Page 191
`
`Enzymatic and
`Chemical
`_Cleavage_ of
`E15100 Proteins
`
`16.4.10
`
`Supplement 28
`
`

`
`The fusion protein must be (at least) partially purified prior to digestion with entero—
`kinase because the enzyme is inactive in crude bacterial lysates.
`
`2. Stop the reaction by adding 30 ul of 2x SDS sample buffer to each reaction. Boil 10
`min.
`
`For larger-scale applications, the reaction can be stopped by adding p-aminobenza-
`midine (PABA) to 5 mM. PABA is a competitive inhibitor of most intestinal serine
`proteases. It should provide protection from nonspecific proteolysis of the reaction
`products by contaminants in the enzyme preparation before the protein of interest is
`purifiedfiuthen
`
`3. Load 10 pl of each sample onto an SDS-polyacrylamide gel to analyze the extent of
`cleavage. Adjust enterokinase concentration and length of incubation accordingly to
`accomplish complete digestion.
`
`4. Scale up the reaction components linearly to digest a larger amount of fusion protein.
`
`Calcium ions marginally increase the efficiency of cleavage, but their presence
`sometimes promotes fusion protein degradation by stimulating contaminating pro-
`teolytic activities. If degradation of the cleaved fusion protein occurs, omit calcium
`and add 5 mM EDTA to the cleavage reaction to try to eliminate the problem.
`
`CHEMICAL CLEAVAGE OF FUSION PROTEINS USING CYANOGEN
`BROMIDE
`
`BASIC
`PROTOCOL 2
`
`Cyanogen bromide (CNBr) has been used to cleave proteins at methionine residues for
`many years. CNBr has been used industrially for the production of both somatostatin
`(Itakura et al., 1977) and insulin (Chance et al., 1981). The reaction is typically carried
`out at low pH in 70% formic acid, and cleavage occurs at the C-terminal side of methionine
`residues. The protein concentration is relatively unimportant, as the CNBr is in vast excess
`for hydrolysis. The technique is useful only if the protein of interest lacks methionine
`residues. Cleavage with CNBr is usually efficient, but side chain modifications and
`nonspecific cleavages are common upon prolonged incubation at low pH. These prob-
`lems, along with the potential for reduction of intramolecular disulfide bonds during
`treatment with 70% formic acid, can be minimized by replacing the formic acid with 6
`M guanidine-HCI/0.2 M HCl.
`
`CAUTION: Cyanogen bromide is extremely toxic. It should only be used in a properly
`ventilated fume hood. Exercise appropriate caution in its use and disposal.
`
`Materials
`
`For recipes, see Reagents and Solutions in this unit (or cross-referenced unit); for common stock
`solutions, see APPENDIX 2; for suppliers, see APPENDIX 4.
`
`1 mg/ml fusion protein
`50 mg/ml cyanogen bromide (CNBr)/70% (v/v) formic acid
`70% (v/v) formic acid
`1x SDS sample buffer (UNI? 10.2)
`
`Additional reagents and equipment for SDS—PAGE (UNIT 10.2)
`
`1. Perform a pilot experiment to determine minimum incubation time. Lyophilize two
`50-ul aliquots of fusion protein solution. Resuspend one aliquot in 50 ul of 50 mg/ml
`CNBr/70% formic acid. Resuspend the other in 50 1.11 of 70% formic acid without
`CNBr. Incubate at room temperature.
`
`2. At 0, 8, 24, and 48 hr, remove 21 5-111 aliquot and lyophilize.
`
`Current Protocols in Molecular Biology
`
`Protein
`Expression
`
`16.4.11
`
`Supplement 28
`
`BEQ 1016
`Page 192
`
`

`
`3. Resuspend all aliquots in 20 |.L1 of 1x SDS sample buffer, boil 10 min, and load onto
`an SDS-polyacrylamide gel.
`
`4. Based on analysis of the gel, determine the minimum incubation time necessary to
`completely cleave the protein.
`
`The protocol can be easily scaled up to accommodate larger amounts offusion protein.
`Some proteins are resistant to cleavage with cyanogen bromide. In such cases, or when
`the fitsion protein to be cleaved is insoluble, guanidine-HCl can be added to the
`reaction at afinal concentration of 6 M.
`
`ALTERNATE
`PROTOCOL 4
`
`CHEMICAL CLEAVAGE OF FUSION PROTEINS USING
`HYDROXLAMINE
`
`Hydroxylamine cleaves proteins at Asn—G1y bonds and can be used as a reagent for
`chemical cleavage of fusion proteins. This cleavage site is less common than that for
`cyanogen bromide, and therefore the presence of a susceptible bond in the protein of
`interest is less likely. One disadvantage is that the released carboxy-terminal fusion partner
`will retain a glycine residue at its amino terminus, which is unacceptable in some
`applications. Also, the reaction requires incubation of the fusion protein at alkaline pH,
`which may cause modification of some amino acid side chains. Finally, protein digestions
`by this technique are usually incomplete due to the nature of the cleavage mechanism
`(E.L., unpub. observ.), reducing yield and possibly complicating post-cleavage purifica-
`tion of the desired protein product. However, the technique does have advantages: speed,
`economy, and the ability to perform digestions under denaturing conditions (e.g., 6 M
`guanidine-HCl) for otherwise insoluble fusion proteins.
`
`CAUTION: Hydroxylamine is potentially explosive if mishandled. Be sure to follow all
`precautions indicated by the manufacturer.
`
`Additional Materials
`
`For recipes, see Reagents and Solutions in this unit (or cross-referenced unit); for common stock
`solutions, see APPENDIX 2; for suppliers, see APPENDIX 4.
`
`1 mg/ml fusion protein in 10 mM Tris-Cl (pH 8.0)/150 mM NaCl
`2x hydroxylamine cleavage solution (see recipe)
`Guanidine-HCl (optional)
`2x SDS sample buffer (UNIT 10.2)
`Boiling water bath
`
`1. Perform a pilot experiment to determine minimum incubation time. Mix 50 ul of 1
`mg/ml fusion protein in 10 mM Tris-Cl (pH 8.0)/150 mM NaCl with 50 pl of 2x
`hydroxylamine cleavage solution in a 1.5-ml microcentrifuge tube. Incubate at 45°C.
`
`Ifthefusion protein is insoluble, guanidine»HCl can be added to the cleavage reaction
`at afinal concentration of6 M. This may also help in cases where a particular Asn-Gly
`bond appears to be resistant to cleavage.
`
`2. At 0, 2, 4, 8, 16, and 24 hr, remove 10-ul aliquots from the cleavage reaction and mix
`with 10 ul of 2x SDS sample buffer. Freeze each tube on dry ice until all time points
`have been collected.
`
`4. Heat samples 10 min in a boiling water bath. Load all samples onto an SDS-poly-
`acrylamide gel to analyze the extent of cleavage.
`
`5. Determine the minimum incubation time necessary for maximum cleavage.
`I_f cleavage after 24 hr is still poor, add guanidine-HCl to 6 M final,
`hydroxylamine concentration to 3 Mfinal, or both.
`
`increase
`
`Cunent Protocols in Molecular Biology
`BEQ 1016
`Page 193
`
`Enzymatic and
`ClEzi1ve::glecta>l'
`Fusion Pmteins
`
`16.4.12
`
`Supplement 28
`
`

`
`CHEMICAL CLEAVAGE OF FUSION PROTEINS BY HYDROLYSIS
`
`AT Low pH
`
`ALTERNATE
`PROTOCOL 5
`
`This method exploits the fact that the Asp-Pro bond is labile at low pH. Hydrolysis of this
`peptide bond occurs at elevated temperatures (37° to 40°C) under acidic conditions (pH
`2.5). Nonspecific cleavages can occur upon prolonged incubation under these conditions,
`and it is necessary to determine empirically the minimum length of time necessary for
`cleavage. Like the other chemical cleavage methods described in this unit, the reaction
`conditions are somewhat harsh and may result in denaturation or modification of the
`protein. On the other hand, this treatment allows insoluble fusion proteins to be cleaved
`by acid hydrolysis of Asp-Pro bonds in the presence of 6 M guanidine-HCl. To use this
`procedure the amino acid sequence of the carboxy-terminal fusion partner should first be
`examined carefully to verify the absence of other Asp-Pro bonds. The released protein
`will retain a proline residue at its amino-terminus.
`
`GST
`This method will potentially cleave any protein containing an Asp-Pro bond.
`fusion vector pGEX1 (UNITI6. 7, and Fig. 16.7.1 therein) contains an Asp-Pro cleavage site
`encoded by the BamHI site of the polylinker cloning region, so fusions at tElS site will
`result in fusion proteins that can be released from GST using this protocol.
`
`Additional Materials
`
`For recipes, see Reagents and Solutions in this unit (or cross-referenced unit); for common stock
`solutions, see APPENDIX 2; for suppliers, see APPENDIX 4.
`
`Fusion protein containing an Asp-Pro bond between the component domains
`70% (v/v) formic acid
`13% (v/v) acetic acid
`0.1 M Tris base
`Guanidine-HCl
`
`1. Perform a pilot experiment to determine optimal hydrolysis conditions. Prepare four
`reaction mixtures:
`
`Reaction 1: ~20 ug fusion protein in 70% formic acid
`
`Reaction 2: ~20 ug fusion protein in 70% formic acid/6 M guanidine-HCI
`
`Reaction 3: ~20 ug fusion protein in 13% acetic acid
`
`Reaction 4: ~20 ug fusion protein in 13% acetic acid/6 M guanidine-HCl.
`
`Incubate all samples at 37°C.
`
`2. At 0, 24, 48, and 72 hr, remove a 5-ug aliquot of each reaction mixture and lyophilize
`to dryness.
`
`The 0 time point can serve as the negative control.
`
`3. Resuspend the hydrolyzed protein in 20 ul of 1x SDS sample buffer and neutralize
`by gradual addition of 0.1 M Tris base until the sample turns from yellow to blue.
`Analyze samples on a tricine SDS-polyacrylamide gel for extent of digestion.
`
`4. Choose the mildest condition and shortest incubation time that give the desired extent
`of cleavage. Scale up to larger amounts of fusion protein accordingly.
`
`Ftere is a great deal of variation in the susceptibility ofAsp-Pro bonds to cleavage.
`Some Asp-Pro bonds cleave readily undermild conditions, whereas others are resistant
`to cleavage and require incubation in stronger acid conditions and/or strong denatur-
`ants to attain hydrolysis. Even under strong conditions, some Asp-Pro bonds remain
`uncleaved and others may not be cleaved to completion, i.e., they may be cleaved in
`only a fraction of the proteins. However; to avoid unwanted denaturation or modifica-
`tion of the protein of interest, it is important to determine the mildest conditions that
`give the desired degree of cleavage.
`
`Cl.ll”:‘61'It Protocols in Molecular Biology
`
`Protein
`Expression
`
`16.4.13
`
`Supplement 28
`
`BEQ 1016
`Page 194
`
`

`
`REAGENTS AND SOLUTIONS
`
`Use deionized, distilled water in all recipes and protocol steps. For common stock solutions, see
`APPENDIX 2; for suppliers, see APPENDIX 4.
`
`Hydroxylamine cleavage solution, 2x
`4 M hydroxylarnine
`0.4 M CHES buffer
`
`Adjust pH to 9.5 with NaOH
`Prepare fresh
`
`Thrombin cleavage buffer
`50 mM Tris-Cl, pH 7.5
`150 mM NaCl
`
`2.5 mM CaCl2
`Store indefinitely at —20°C
`
`COMMENTARY
`
`Background Information
`The use of fusion proteins in Escherichia
`coli for production of proteins from other or-
`ganisms is becoming increasingly popular. The
`principal advantages of fusion proteins include
`high expression levels, ease ofpurification, and
`ease of detection with biochemical or im-
`
`munological reagents. However, early protein
`fusion methods fused the target protein to the
`carboxy-terminus of E. coli B—ga1actosidase;
`such proteins are usually insoluble, have re-
`stricted usefulness (except as antigens for an-
`tibody pE'0ClllCtlOl'l), and require refolding of the
`protein in an attempt to retain biological activ-
`ity. The development of improved fusion pro-
`tein expression systems that are often capable
`of producing properly folded and biologically
`active proteins in E. coli, such as the MBP
`system (wwr 16.6), the GST system (wwr 16.7),
`and the Trx system (UNITI6.8), has increased the
`need for suitable methods of separating the
`protein of interest from the so—called carréer
`protein. Such separation is desirable only after
`the carrier domain has been exploited for its
`particular attributes—e.g, to facilitate specific
`purification or detection, or to enhance stability
`or solubility. Subsequent site-specific cleavage
`of the carrier domain from the correctly folded
`protein of interest then allows evaluation of the
`biological activity of the protein without poten-
`tial interference from a covalently-attached fu-
`sion partner.
`Over the years, many different enzymatic
`and chemical methods for site-specific cleav-
`age of polypeptides have been developed
`(Gross, 1967; Spande et al., 1970; Maroux et
`al., 1971; Landon, 1977; Nagai and Thtzigersen,
`1984; Chang, 1985; Szoka et al., 1986; Smith
`and Johnson, 1988; Gearing, et al., 1989; Ham-
`
`Enzymatic and
`Chemical
`Cleavage of
`Fusion Proteins
`
`16.4.14
`
`Supplement 28
`
`mond et al., 1991). The choice of cleavage
`reagents for those expression vectors obtained
`from molecular biology suppliers that are used
`“off the shelf’ will usually be dictated by the
`recognition sequences that lie at their respec-
`tive fusion junctions. For example, MBP vec-
`tors (UNIT 16.6; Maina et al., 1988) are designed
`for factor Xa cleavage, GST vectors (wvrr 16.7)
`offer a choice of factor Xa, thrombin, or acid
`cleavage, and the Trx vector (UNlTI6.8; LaVa1lie
`et al. , 1993a) utilizes enterolcinase for cleavage.
`However, it is importantto remember that these
`vectors, or any other fusion protein expression
`vector, can be manipulated by the user to in-
`clude junction amino acids that will allow en-
`zymatic or chemical hydrolysis with other
`cleavage reagents.
`
`Critical Parameters and
`
`Troubleshooting
`The cleavage methods described in this unit
`have been chosen based upon their specificity,
`efficiency, and reagent availability. Uitimately,
`the choice of cleavage reagent will depend
`upon many factors. First of all, the primary
`sequence of the protein of interest must be
`scrutinized to identify sequences that will be
`susceptible to the cleavage reagents in ques-
`tion. Because of the highly restricted specific-
`ity in the case of the proteases described in the
`preceding protocols, the occurrence of such
`additional sites is unlikely. However, the use of
`chemical cleavage methods or less specific
`proteases such as trypsin (Lysi or Argi) re-
`quires careful consideration of the composition
`of the protein of interest to avoid unwanted
`fragmentation of the product. Secondly, the
`physical characteristics of the fusion protein
`are an important consideration in choosing an
`
`Current Protocols in Molecular Biology
`BEQ 1016
`Page 195
`
`

`
`appropriate cleavage method. Fusion proteins
`that are insoluble generally require a chemical
`cleavage method that allows incubation in the
`presence of protein denaturants such as
`guanidine-HCl or urea followed by proper re-
`folding. Conversely, soluble fusion proteins
`should be cleaved under conditions where de-
`
`naturation is minimized to preserve their struc-
`ture and/or biological activity. In addition,
`some cleavage methods require incubation at
`pH extremes, which may result in modifica-
`tions to some of the side chains or aggregation
`and precipitation of the protein and may ulti-
`mately be deleterious to the usefulness of the
`cleaved product.
`When the fusion protein is produced in a
`soluble fashion, enzymatic cleavage protocols
`are preferred for many reasons. These reactions
`are carried out under mild conditions of neu1ral
`
`pH, low ionic strength, and moderate temper-
`ature (25° to 37°C). Such conditions approxi-
`mate physiological environments and typically
`should be least harmful to the integrity of the
`protein. The high degree of specificity exhib-
`ited by these proteases ensures a low probabil-
`ity of unwanted cleavage elsewhere in the pro-
`tein of interest. Finally, the extent of enzymatic
`cleavage typically can be altered by modulat-
`ing parameters such as amount of enzyme,
`subsfzate concentration, and length of incuba-
`tion.
`
`The use of proteases sometimes causes
`problems, however. The most common prob-
`lem is unwanted secondary proteolysis or deg-
`radation of the fusic:-n protein by contaminating
`proteolytic activities in the cleavage reaction.
`Degradation may be caused by E. cali proteases
`that have not been purified away from the
`fusion protein prior to cleavage. Such contam-
`inants can often be alleviated by additional
`purification prior to enzymatic digestion. Al-
`ternatively, the unwanted proteolytic activéty
`may result from a contaminant in the enzyme
`preparation itself. These enzymes are serine
`proteases that have been purified from natural
`sources, so it is probable that they are contam-
`inated with other proteases that copurify in
`trace amounts. For example, even highly puri-
`fied enterokinase from bovine intestine con-
`
`tains Irace amounts of tryptic and chymotryptic
`activity that can cause minor secondary prote-
`olysis of thioredoxin fusion proteins (LaVallie
`et al., 1993a). This degradation can be reduced
`by omitting Ca” ions from the digestion and
`using EDTA to chelate residual Ca”. The best
`solution to this problem, however, is to use a
`recombinant source of enzyme produced in cell
`
`Current Protocols in Molecular Biology
`
`culture; such enzymes are free of contaminat-
`ing proteases found in intesténal preparations
`(LaVallie et al., 1993b and unpub. observ.).
`Another potential problem that has been
`observed with some enzymatic cleavages is
`cleavage at sites other than the anticipated
`peptide bonds. This has been reported for factor
`Xa (Nagai and Thgzsgersen, 1987; Lauritzen et
`al., 1991) and thrombin (Chang, 1985); entero—
`kinase has been observed to cleave at subsites
`
`that resemble the (Asp)4-Lys recognition se-
`quence when the substrate is denatured or oth-
`erwise improperly folded (Light et al., 1980, E.
`LaVallie and L. Racie, unpub. observ.). Almost
`always, this relaxed site specificity can be min-
`imized by decreasing the enzyme/substrate
`ratio and/or the éme of incubation. There are
`
`also more exotic strategies; for example, re-
`versible acylation of the fusion protein has been
`used to eliminate nonspecific cleavage by fac-
`tor Xa (Wearne, 1990).
`Although it is less desirable than enzymatic
`digestion in most circumstances, chemical
`cleavage of fusion proteins is sometimes nec-
`essary and may be advantageous in certain
`applications. This is in spite of the significant
`disadvantages. First, chemical cleavage proce-
`dures almost universally employ harsh condi-
`tions, such as pH extremes or high tempera-
`tures,
`that can denature the fusion protein
`and/or modify amino acid side chains (e.g.,
`deamidation of Asn residues and oxidation of
`
`Met residues). Second, cleavage specifscity
`tends to be limited to single amino acids or, at
`most, dipeptide sequences, greatly decreasing
`the utility of chemical reagents in site—specific
`cleavage of large polypeptide substrates. Even
`this low degree of specificity is not absolute,
`and low levels of side reactions at alternate sites
`
`have been noted for hydroxylamine (Bornstein
`and Balian, 1970; Steinman et al., 1974) and
`cyanogen bromide (Langley and Smith, 1971).
`Even acidic cleavage of Asp—Pro bonds is
`sometimes accompanied by nonspecific pep-
`tide bond hydrolysis when incubation is pro-
`longed (Landon, 1977). Third, using chemical
`cleavage reagents involves the danger of work-
`ing with hazardous compounds such as cyano-
`gen bromide and hydroxylamine. Ex1Ieme care
`must be used in the storage, use, and disposal
`of these reagents. Material Safety Data Sheets
`(MSDSs) should be obtained from the manu-
`facturer for any of these compounds and should
`be read carefully.
`In spite of these shortcomings, however,
`specific chemical hydrolysis of junction pep-
`tide bonds in fusion proteins is preferred for
`
`Protein
`Expression
`
`16.4.15
`
`Supplement 28
`
`BEQ 1016
`Page 196
`
`

`
`some applications. For instance, fusion pro-
`teins that are produced in an insoluble form
`cannot be cleaved enzymatically unless they
`have been solubilized and refolded, but site-
`
`specific cleavage of the protein domains often
`can be accomplished using chemical hydroly-
`sis in the presence of strong chaotropic agents
`such as guanidine-HCl (Landon, 1977; Szoka
`et al., 1986; Villa et al., 1989). Chemical cleav-
`ages are also useful when a soluble fusion pro-
`tein is refractory to cleavage under nondenatur-
`ing conditions using either enzymatic or chem-
`ical cleavage methods. Other advantages of
`chemical cleavage reagents are economy, pu-
`rity, and wide availability.
`
`Anticipated Results
`Enzymatic digestion of fusion proteins is
`sensitive to many different parameters such as
`temperature, pH, ionic strength, buffer compo-
`sition, substaate concentration, enzyme con-
`centration, and length of incubation. Optimum
`parameters must be deterznined empirically for
`each fusion protein. However, if the fusion
`protein is of adequate purity and the cleavage
`site is accessible, in most cases the standard
`conditions described in the protocols will give
`satisfactory cleavage.
`Chemical cleavages are much more forgiv-
`ing of small variations in reaction conditions,
`and cleavage should be attainable using the
`reaction conditions given. However, the extent
`of protein modifications caused by these gen-
`eral reaction conditions may often be signifi-
`cant to the ultimate usefulness of the cleaved
`
`protein. Reaction parameters may be adjusted
`as described to minimize any modifications or
`secondary cleavages that may occur.
`With any of these methods, complete diges-
`tion of the fusion protein is often difficult to
`attain, and cleavage efficiencies of 70% to 80%
`should be considered satisfactory.
`
`Time Considerations
`
`Fusion protein cleavage can be accom-
`plished in hours or days, depending upon the
`reagents used and the reaction conditions. For
`factor Xa, thrombin, and enterokinase, the en-
`zyme—to-substrate ratio and substrate concen-
`tration are often chosen so the digestions are
`usually complete in 8 to 24 hr. By contrast,
`chemical cleavages are often highly variable in
`the amount oftime necessary for total cleavage.
`Overnight digestion under standard conditions
`is common for cyanogen bromide and acid
`cleavages, whereas hydroxylamine cleavages
`are often complete in 2 to 4 hr.
`
`Literature Cited
`Bomstein, P. and Balian, G. 1970. The specific
`nonenzymatic cleavage of bovine ribonuclease
`with hydroxylamine. J. Biol. Chem. 245:4854-
`4856.
`
`Bomstein, P. and Balian, G. 1977. Cleavage at Asn-
`Gly bonds with hydroxylarnine. Methods En-
`zymol. 47: 132-145.
`Chance, R.E., Hoffman, J.A., Kroeff, E.P., Johnson,
`M.G., Schirmer, E.W., Bromer, W.W., Ross,
`M.J., and Wetzel, R. 1981. The production of
`human insulin using recombinant DNA technol-
`ogy and a new chain combination procedure. In
`Peptides: Synthesis-Structure-Function (D.H.
`Rich and E. Gross, eds.), pp. 721-728. Pierce
`Chemical, Rockford, Ill.
`
`Chang, J.-Y. 1985. Thrombin specificity. Eur. J.
`Biochem. 15l:217-224.
`
`Gearing, D.P., Nicola, N.A., Metcalf, D., Foote, S.,
`Willson, T.A., Gough, N.M., and Williams, R.L.
`1989. Production of leukemia factor in Es-
`cherichia coli by a novel procedure and its use
`in maintaining embryonic stem cells in culture.
`Bio/Technology 7: 1 157-1161.
`
`Gross, E. 1967. The cyanogen bromide reaction.
`Methods Enzymol. 1 1 1238-255.
`Hammond, P.M., Atkinson, T., Sherwood, R.F., and
`Scawen, M.D. 1991. Manufacturing new-gener-
`ation proteins. Part I: The technology. BioPharm.
`4: 16-23.
`
`ltakura, K., Hirose, T., Crea, R., Riggs, A.D., Heyne-
`ker, H.L., Bolivar, F., and Boyer, H.W. 1977.
`Expression in Escherichia coli of a chemically
`synthesized gene for the hormone somatostatin.
`Science l98:1056-1063.
`
`Langley, T]. and Smith, E.L. 1971. Sequence of
`bovine liver glutamate dehydrogenase. IV. Cy-
`anogen bromide peptides. J. Biol. Chem.
`2746:3789-3801.
`
`Landon, M. 1977. Cleavage at aspartyl-prolyl
`bonds. Methods Enzymol. 47(E): 145-149.
`Lauritzen, C., Tiichsen, E., Hansen, P.E., and
`Skovgaard, O. 1991. BPTI and N-terminal ex-
`tended analogues generated by Factor Xa cleav-
`age and cathepsin C trimming of a fusion protein
`expressed in Escherichia coli. Pnot. Expr. Pursf
`2: 372-378.
`
`LaVa1lie, E.R., DiBlasio, E.A., Kovacic, S., Grant,
`K.L., Schendel, RE, and McCoy, J .M. 1993a. A
`thioredoxin gene fusion system that circumvents
`inclusion body formation in the E. coli cyto-
`plasm. Bio/Technalogy 11:187-193.
`LaVallie, E.R., Rehemtulla, A., Racie, L.A.,
`DiBlasio, E.A., Ferenz, C., Grant, K.L., Light,
`A., and McCoy, J.M. 1993b. Cloning and func-
`tional expression of a cDNA encoding the
`catalytic subunit of bovine enterokinase. J. Biol.
`Chem. 268:23311-23317.
`
`Light, A., Savithri, H. S., and Liepnieks, J.J. 1980.
`Specificity of bovine enterokinase toward pro-
`tein substrates. Anal. Biochem. 106: 199-206.
`
`Current Protocols in Molecular Biology
`BEQ 1016
`Page 197
`
`Enzymatic and
`Chemical
`Cleavage of
`Fusion Proteins
`
`16.4.16
`
`Supplement 28
`
`

`
`Maina, C.V., Riggs, P.D., Grandea, A.G., Slatko,
`B.E., Moran, L.S., Tagliamonte, J.A., McRey—
`nolds, L.A., and Guan, C. 1988. An Escherichia
`coli vector to express and purify foreigr: proteins
`by fusion to and separation from maltose-bind-
`ing protein. Gene 74:365-373.
`Maroux, S., Baratti, J., and Desnuelle, P. 1971.
`Purification and specificity of porcine enteroki-
`nase. .1. Biol. Chem. 246:5031-5039.
`
`Nagai, K. and Thogersen, H. C. 1984. Generation of
`B-globin by sequence-specific proteoiysis of a
`hybrid protein in Escherichia coli. Nature
`309:810-812.
`
`Nagai, K. and Thogersen, H. C. 1987. Synthesis and
`sequence-specific proteolysis of hybrid proteins
`produced in Escherichia coli. Methods EnzymoL
`153:461—481.
`
`Smith, D.B. and Johnson, K.S. 1988. Single-step
`purification of polypeptides expressed in Es-
`cherichia coli as fusions with glutathione S-
`transferase. Gene 67:31-40.
`
`Spande, T.F., Witkop, 13., Degani, Y., and Pat-
`chomik, A. 1970. Selective cleavage and modi—
`fication of peptides and proteins. Adv. Prot.
`Chem. 24:97—260.
`
`Steinman, H.M., Naik, V.R., Abemethy, J.L., and
`Hill, R.L. 1974. Bovine erythrocyte superoxide
`dismutase: Complete amino acid sequence. J.
`Biol. Chem. 249:7326-7338.
`
`Szoka, P.R., Schreiber, A.B., Chan, H., and Murthy,
`J . 1986. A general method for retrieving compo—
`nents of a genetically engineered fusion protein.
`DNA 5:11-20.
`
`Villa, S., DeFazio, G., and Canosi, U. 1989. Cyano-
`gen bromide cleavage at methionine residues of
`polypeptides containing disulfide bonds. Anal.
`Biochem. 177:161-164.
`
`Wearne, S.J. 1990. Factor Xa cleavage of fusion
`proteins: Elimination of nonspecific cleavage by
`reversible acylation. FEBS Lett. 263:23—26.
`
`Contributed by Edward R. LaVallie
`and John M. McCoy
`Genetics Institute
`
`Cambridge, Massachusetts
`
`Donald B. Smith (throirebin cleavage)
`University of Edinburgh
`Edinburgh, Scotland
`
`Paul Riggs (denaturing fusion proteins)
`New England Biolabs
`Beverly, Massachusetts
`
`Current Protocols in Molecular Biology
`
`Protein
`Expression
`
`16.4.17
`
`Supplement 28
`
`BEQ 1016
`Page 198
`
`

`
`UNIT 16.5 Expression and Purification of lacZ and
`trpE

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket