13
Engineering Kunitz Domain 1 (KD1) of Human Tissue Factor Pathway Inhibitor-2 to Selectively Inhibit Fibrinolysis PROPERTIES OF KD1-L17R VARIANT * S Received for publication, October 3, 2010, and in revised form, November 17, 2010 Published, JBC Papers in Press, November 29, 2010, DOI 10.1074/jbc.M110.191163 Madhu S. Bajaj ‡1 , Godwin I. Ogueli § , Yogesh Kumar § , Kanagasabai Vadivel § , Gregory Lawson , Sreejesh Shanker §2 , Amy E. Schmidt §3 , and S. Paul Bajaj §4 From the Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, § Protein Chemistry Laboratory, UCLA/Orthopaedic Hospital, Department of Orthopaedic Surgery and Molecular Biology Institute, UCLA, and the Division of Laboratory Animal Medicine, UCLA, Los Angeles, California 90095 Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly, it has been proposed for use as an anticoagulant. Full- length TFPI-2 or its isolated first Kunitz domain (KD1) also inhibits plasmin; therefore, it has been proposed for use as an antifibrinolytic agent. However, the anticoagulant prop- erties of TFPI-2 or KD1 would diminish its antifibrinolytic function. In this study, structure-based investigations and analysis of the serine protease profiles revealed that coagu- lation enzymes prefer a hydrophobic residue at the P2 posi- tion in their substrates/inhibitors, whereas plasmin prefers a positively charged arginine residue at the corresponding position in its substrates/inhibitors. Based upon this obser- vation, we changed the P2 residue Leu-17 in KD1 to Arg (KD1-L17R) and compared its inhibitory properties with wild-type KD1 (KD1-WT). Both WT and KD1-L17R were expressed in Escherichia coli, folded, and purified to homo- geneity. N-terminal sequences and mass spectra confirmed proper expression of KD1-WT and KD1-L17R. Compared with KD1-WT, the KD1-L17R did not inhibit factor XIa, plasma kallikrein, or factor VIIa/tissue factor. Furthermore, KD1-L17R inhibited plasmin with 6-fold increased affin- ity and effectively prevented plasma clot fibrinolysis in- duced by tissue plasminogen activator. Similarly, in a mouse liver laceration bleeding model, KD1-L17R was 8-fold more effective than KD1-WT in preventing blood loss. Im- portantly, in this bleeding model, KD1-L17R was equally or more effective than aprotinin or tranexamic acid, which have been used as antifibrinolytic agents to prevent blood loss during major surgery/trauma. Furthermore, as com- pared with aprotinin, renal toxicity was not observed with KD1-L17R. Serine proteases are ubiquitous in living organisms and are involved in many physiologic processes, including digestion and respiration (1–3), blood coagulation and fibrinolysis (4, 5), kinin formation and tumorigenesis (6), complement acti- vation and phagocytosis (7), osteoarthritis and bone remodel- ing (8, 9), as well as in ovogenesis and fertilization (10). Bio- logic activity of serine proteases is tightly regulated by their cognate inhibitors. A majority of these inhibitors are proteins, and based upon their unique folding characteristics, they are classified into the Kunitz (11), Kazal (12), Serpin (13), and mucus (14) families. The prototypical Kunitz inhibitor, apro- tinin, also referred to as bovine basic pancreatic trypsin inhib- itor (BPTI), 5 is a single chain protein of 58 amino acids and has three intrachain disulfide bonds that are conserved in all family members (15). Although a human homolog of BPTI has not been identified, several larger human proteins that contain one or more Kunitz inhibitory domains are known. Kunitz domain-containing proteins that inhibit coagulation and/or fibrinolysis include tissue factor pathway inhibitor type-1 (TFPI) (16), tissue factor pathway inhibitor type-2 (TFPI-2) (17), bikunin (18), and membrane-associated amy- loid -precursor protein (APP) (19). TFPI contains three Kunitz-type domains, and the physiologic function of the N- terminal first domain is to inhibit factor VIIa/tissue factor (FVIIa/TF), whereas the function of the second domain is to inhibit factor Xa (16). However, the isolated first domain also inhibits plasmin (Pm) and cathepsin G, whereas the isolated second domain also inhibits trypsin and chymotrypsin (20). No direct protease-inhibiting function has been ascribed to the third Kunitz domain (21). TFPI-2 features a domain orga- nization similar to TFPI and via its Kunitz domain-1 inhibits * This work was supported, in whole or in part, by National Institutes of Health Grants R21HL89661 and RO1HL36365. This article was selected as a Paper of the Week. S The on-line version of this article (available at http://www.jbc.org) con- tains supplemental Fig. 1S. 1 To whom correspondence may be addressed: Medicine-Pulmonary Critical Care, David Geffen School of Medicine, UCLA, Box 951690, Los Angeles, CA 90095-1690. Tel.: 310-825-8024; Fax: 310-825-5972; E-mail: [email protected]. 2 Present address: Dept. of Biochemistry and Molecular Biology, Baylor Col- lege of Medicine, Houston, TX 77030. 3 Present address: Dept. of Pathology and Immunology, Washington Uni- versity School of Medicine, St. Louis, MO 63110. 4 To whom correspondence may be addressed: Protein Chemistry Labora- tory, Dept. of Orthopaedic Surgery and Molecular Biology Institute, UCLA/Orthopaedic Hospital, Los Angeles, CA, 90095-1795. E-mail: [email protected]. 5 The abbreviations used are: BPTI, bovine pancreatic trypsin inhibitor; TFPI, tissue factor pathway inhibitor-1; TFPI-2, tissue factor pathway inhibi- tor-2; KD1, Kunitz domain 1 of TFPI-2; APP, amyloid -precursor protein; IIa, -thrombin; factor VIIa, FVIIa; TF, tissue factor; sTM, soluble thrombo- modulin; sTF, soluble TF; FXIa, factor XIa; APC, activated protein C; tPA, tissue plasminogen activator; pKLK, plasma kallikrein; Pm, plasmin; aPTT, activated partial thromboplastin time; TE, tranexamic acid; NPP, normal pooled plasma. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 6, pp. 4329 –4340, February 11, 2011 © 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4329 by guest on August 20, 2019 http://www.jbc.org/ Downloaded from

EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

Embed Size (px)

Citation preview

Page 1: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

Engineering Kunitz Domain 1 (KD1) of Human Tissue FactorPathway Inhibitor-2 to Selectively Inhibit FibrinolysisPROPERTIES OF KD1-L17R VARIANT*□S �

Received for publication, October 3, 2010, and in revised form, November 17, 2010 Published, JBC Papers in Press, November 29, 2010, DOI 10.1074/jbc.M110.191163

Madhu S. Bajaj‡1, Godwin I. Ogueli§, Yogesh Kumar§, Kanagasabai Vadivel§, Gregory Lawson¶, Sreejesh Shanker§2,Amy E. Schmidt§3, and S. Paul Bajaj§4

From the ‡Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA,§Protein Chemistry Laboratory, UCLA/Orthopaedic Hospital, Department of Orthopaedic Surgery and Molecular Biology Institute,UCLA, and the ¶Division of Laboratory Animal Medicine, UCLA, Los Angeles, California 90095

Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factorXIa, plasma kallikrein, and factor VIIa/tissue factor; accord-ingly, it has been proposed for use as an anticoagulant. Full-length TFPI-2 or its isolated first Kunitz domain (KD1) alsoinhibits plasmin; therefore, it has been proposed for use asan antifibrinolytic agent. However, the anticoagulant prop-erties of TFPI-2 or KD1 would diminish its antifibrinolyticfunction. In this study, structure-based investigations andanalysis of the serine protease profiles revealed that coagu-lation enzymes prefer a hydrophobic residue at the P2� posi-tion in their substrates/inhibitors, whereas plasmin prefersa positively charged arginine residue at the correspondingposition in its substrates/inhibitors. Based upon this obser-vation, we changed the P2� residue Leu-17 in KD1 to Arg(KD1-L17R) and compared its inhibitory properties withwild-type KD1 (KD1-WT). Both WT and KD1-L17R wereexpressed in Escherichia coli, folded, and purified to homo-geneity. N-terminal sequences and mass spectra confirmedproper expression of KD1-WT and KD1-L17R. Comparedwith KD1-WT, the KD1-L17R did not inhibit factor XIa,plasma kallikrein, or factor VIIa/tissue factor. Furthermore,KD1-L17R inhibited plasmin with �6-fold increased affin-ity and effectively prevented plasma clot fibrinolysis in-duced by tissue plasminogen activator. Similarly, in a mouseliver laceration bleeding model, KD1-L17R was �8-foldmore effective than KD1-WT in preventing blood loss. Im-portantly, in this bleeding model, KD1-L17R was equally ormore effective than aprotinin or tranexamic acid, whichhave been used as antifibrinolytic agents to prevent blood

loss during major surgery/trauma. Furthermore, as com-pared with aprotinin, renal toxicity was not observed withKD1-L17R.

Serine proteases are ubiquitous in living organisms and areinvolved in many physiologic processes, including digestionand respiration (1–3), blood coagulation and fibrinolysis (4,5), kinin formation and tumorigenesis (6), complement acti-vation and phagocytosis (7), osteoarthritis and bone remodel-ing (8, 9), as well as in ovogenesis and fertilization (10). Bio-logic activity of serine proteases is tightly regulated by theircognate inhibitors. A majority of these inhibitors are proteins,and based upon their unique folding characteristics, they areclassified into the Kunitz (11), Kazal (12), Serpin (13), andmucus (14) families. The prototypical Kunitz inhibitor, apro-tinin, also referred to as bovine basic pancreatic trypsin inhib-itor (BPTI),5 is a single chain protein of 58 amino acids andhas three intrachain disulfide bonds that are conserved in allfamily members (15). Although a human homolog of BPTIhas not been identified, several larger human proteins thatcontain one or more Kunitz inhibitory domains are known.Kunitz domain-containing proteins that inhibit coagulation

and/or fibrinolysis include tissue factor pathway inhibitortype-1 (TFPI) (16), tissue factor pathway inhibitor type-2(TFPI-2) (17), bikunin (18), and membrane-associated amy-loid �-precursor protein (A�PP) (19). TFPI contains threeKunitz-type domains, and the physiologic function of the N-terminal first domain is to inhibit factor VIIa/tissue factor(FVIIa/TF), whereas the function of the second domain is toinhibit factor Xa (16). However, the isolated first domain alsoinhibits plasmin (Pm) and cathepsin G, whereas the isolatedsecond domain also inhibits trypsin and chymotrypsin (20).No direct protease-inhibiting function has been ascribed tothe third Kunitz domain (21). TFPI-2 features a domain orga-nization similar to TFPI and via its Kunitz domain-1 inhibits

* This work was supported, in whole or in part, by National Institutes ofHealth Grants R21HL89661 and RO1HL36365.

� This article was selected as a Paper of the Week.□S The on-line version of this article (available at http://www.jbc.org) con-

tains supplemental Fig. 1S.1 To whom correspondence may be addressed: Medicine-Pulmonary Critical

Care, David Geffen School of Medicine, UCLA, Box 951690, Los Angeles,CA 90095-1690. Tel.: 310-825-8024; Fax: 310-825-5972; E-mail:[email protected].

2 Present address: Dept. of Biochemistry and Molecular Biology, Baylor Col-lege of Medicine, Houston, TX 77030.

3 Present address: Dept. of Pathology and Immunology, Washington Uni-versity School of Medicine, St. Louis, MO 63110.

4 To whom correspondence may be addressed: Protein Chemistry Labora-tory, Dept. of Orthopaedic Surgery and Molecular Biology Institute,UCLA/Orthopaedic Hospital, Los Angeles, CA, 90095-1795. E-mail:[email protected].

5 The abbreviations used are: BPTI, bovine pancreatic trypsin inhibitor; TFPI,tissue factor pathway inhibitor-1; TFPI-2, tissue factor pathway inhibi-tor-2; KD1, Kunitz domain 1 of TFPI-2; A�PP, amyloid �-precursor protein;IIa, �-thrombin; factor VIIa, FVIIa; TF, tissue factor; sTM, soluble thrombo-modulin; sTF, soluble TF; FXIa, factor XIa; APC, activated protein C; tPA,tissue plasminogen activator; pKLK, plasma kallikrein; Pm, plasmin; aPTT,activated partial thromboplastin time; TE, tranexamic acid; NPP, normalpooled plasma.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 286, NO. 6, pp. 4329 –4340, February 11, 2011© 2011 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4329

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 2: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

trypsin, chymotrypsin, VIIa/TF, factor XIa (FXIa), plasmakallikrein (pKLK), and Pm (22, 23). The other two Kunitz do-mains in TFPI-2 have no known inhibitory function. Placentalbikunin inhibits several proteases involved in intrinsic coagu-lation and fibrinolysis (18), whereas the Kunitz domain ofA�PP inhibits FXIa and Pm (24). Thus, during evolution, in-sertion and/or duplication resulted in a diverse Kunitz familyof proteins containing a broad spectrum of inhibitory as wellas noninhibitory modules.BPTI inhibits several serine proteases and has been used as

a therapeutic agent, Trasylol� (25), to reduce blood loss dur-ing cardiac surgery (26). These benefits appear to be derivedfrom its pKLK and/or Pm inhibitory activity (25). However, itsuse has been linked to kidney damage, myocardial infarction,and strokes (27, 28). Notably, BPTI is of bovine origin, and itsanaphylactic potential has been a major concern (29). Forthese reasons, BPTI (aprotinin) has been taken off the clinicalmarket. Thus, a human homolog with the least modificationsto yield superior biochemical and clinical characteristics isneeded to replace BPTI.Several investigators have used a suitable 58-residue human

Kunitz domain to obtain selective inhibitors of pKLK and/or Pm.Dennis et al. (30) andMarkland et al. (31) used the A�PP do-main (30) or the TFPI domain-1 (31) as a scaffold and phage dis-play technology to obtain potent pKLK inhibitors, termedKALI-DY and EPI-K503, respectively. KALI-DY (30) differed insix residues fromA�PP, and EPI-K503 (31) differed in seven resi-dues fromTFPI. Furthermore, KALI-DY also inhibited FXIa,whereas EPI-K503 inhibited FXIa and Pmwith aKi �30 nM. In aconceptually similar study, Markland et al. (32) used TFPI do-main-1 to generate a potent Pm inhibitor, termed EPI-P302. TheEPI-P302 differed in seven residues fromTFPI domain-1 andexhibited very high affinity for Pm; however, it also inhibitedFXIa and pKLKwithKi �200 nM.Moreover, in each case, asnoted above, several residues were changed from the startingmolecular scaffold, which could elicit an immune response inhuman subjects. In fact, anaphylactic reactions have been ob-served in patients treated with EPI-K503/DX-88 (33).In this study, we used serine protease S2�/P2� subsite6 pro-

filing and a structure-based approach to engineer the Kunitzdomain 1 of TFPI-2 (KD1) to selectively inhibit fibrinolysis.Our analysis indicates that coagulation proteases prefer hy-drophobic residues at the P2� position in their substrates/inhibitors, whereas Pm, a fibrinolytic enzyme, prefers a basicresidue at this position. Thus, we inferred that replacing Leuwith Arg at the P2� site in KD1 could abolish its anticoagulant

property while simultaneously enhancing its antifibrinolyticfunction. Moreover, such a small change in KD1 may notelicit an immune response in humans. Furthermore, as com-pared with BPTI (aprotinin) or tranexamic acid (TE), KD1-L17R was found to be more effective in reducing blood loss ina mouse liver laceration model, and kidney toxicity was notobserved with KD1-L17R.

EXPERIMENTAL PROCEDURES

Materials—Escherichia coli strain BL21(DE3) pLysS andpET28a expression vector were obtained from Novagen Inc.(Madison, WI). Amicon centrifugal filter devices (3000Mrcutoff) were purchased fromMillipore (Bedford, MA). Q-Sepharose FF, Superdex 200, and His-Trap HP columns wereobtained from Amersham Biosciences. Kanamycin and iso-propyl thiogalactopyranoside were obtained from Sigma. Hu-man �-thrombin (IIa), activated protein C (APC), FXIa,pKLK, and Pm were purchased from Hematologic Technolo-gies Inc. BPTI was received from ZymoGenetics (Seattle), andhuman factor VIIa (FVIIa) was prepared as described previ-ously (35). Tissue plasminogen activator (tPA; Alteplase) waspurchased from Genentech (South San Francisco). Recombi-nant sTF was provided by Tom Girard (Monsanto/Pfizer, St.Louis, MO), and soluble thrombomodulin (sTM) was receivedfrom Dr. S. Krishnaswamy (Children’s Hospital of Philadel-phia). Recombinant TF containing the transmembrane do-main (residues 1–243) was a gift from Genentech (San Fran-cisco). Pm substrate S-2251 (H-D-Val-Leu-Lys-p-nitroanilide),pKLK, and FXIa substrate S-2366 (pyroGlu-Pro-Arg-p-nitroa-nilide), IIa and IIa/sTM substrate S-2238 (H-D-Phe-pip-Arg-pnitroanilide), and tPA, APC, and FVIIa substrate S-2288 (H-D-Ile-Pro-Arg-p nitroanilide) were obtained from DiapharmaInc. All other reagents were of the highest purity commer-cially available.Expression and Purification of KD1-WT and KD1-L17R—

Residues 1–73 of human TFPI-2 containing the first Kunitzdomain were cloned and overexpressed as an N-terminalHis6-tagged fusion protein in E. coli strain BL21(DE3) pLysSusing the T7 promoter system. The recombinant plasmid de-rived from pET28a, containing a His6 leader sequence fol-lowed by a thrombin cleavage site and the cDNA encodingthe KD1, was prepared according to standard procedures (36).The point mutant KD1-L17R was generated using theQuikChange� site-directed mutagenesis kit (Stratagene, LaJolla CA) according to the manufacturer’s instructions. Bothrecombinant constructs were checked for in-frame orienta-tion by nucleic acid sequencing. The His6-tagged KD1-WTand KD1-L17R fusion proteins were expressed in E. coligrown in Luria broth containing 15 mg/liter kanamycin andinduced at 37 °C with 1 mM isopropyl thiogalactopyranosideat mid-log phase (A600 �0.9) for 4–6 h at 37 °C. The cellswere harvested by centrifugation at 6000 � g and stored fro-zen at �20 °C.The frozen cell pellets were suspended in 50 mM Tris, pH

7.2, containing a complete Mini� protease inhibitor mixturetablet from Roche Applied Science. After 15 min on ice, thecells were sonicated in lysis buffer (50 mM Tris, 60 mM EDTA,1.5 M NaCl, 6% Triton X-100, pH 7.2), and inclusion bodies

6 KD1 and BPTI residues P2, P1, P1�, P2� are numbered according toSchechter and Berger (34). The corresponding subsites are S2, S1, S1�, S2�in the enzyme, respectively. The residue numbering corresponds to thatof BPTI such that residue 15 is at the P1 position. For comparison, KD1 isnumbered using the BPTI numbering system such that residue 15 in BPTInumbering corresponds to residue 24 in KD1. Thus, the BPTI numberingand KD1 numbering differ by 9 throughout. The chymotrypsin aminoacid numbering system is used throughout for the protease domain ofPm, FXIa, pKLK, and FVIIa. For example, residue 73 in chymotrypsin num-bering corresponds to 623 in Pm, 433 in FXIa, 435 in pKLK, and 213 inFVIIa. In Fig. 7 and Table 2, the amino acid corresponding to a given pro-tease is given in braces (e.g. {623}). Where insertions occur, the chymo-trypsin numbering is followed by a capital letter, such as A.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

4330 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 6 • FEBRUARY 11, 2011

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 3: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

were recovered by centrifugation for 30 min at 12,000 � g.The inclusion bodies were solubilized in 20 mM sodium phos-phate, pH 7.4, containing 0.5 M NaCl, 10 mM �-mercaptoetha-nol, and 8 M urea. The insoluble material was removed bycentrifugation at 10,000 � g for 10 min, and the supernatantwas applied to a nickel-charged His-Trap column. The col-umn was washed with equilibration buffer (20 mM sodiumphosphate, 0.5 M NaCl, 6 M urea, and 25 mM imidazole), andthe His6-tagged KD1-WT (or KD1-L17R) fusion protein waseluted using a 25 mM to 1 M imidazole gradient.

The His-Trap purified KD1-WT (or KD1-L17R) was re-duced with 50 mM dithiothreitol and dialyzed against 20 vol-umes of 50 mM Tris, pH 9.0, 6 M urea for 12 h at 4 °C. Thedialyzed fusion protein was refolded using the reduced andoxidized glutathione system as described earlier (37, 38). Therefolded protein was then purified using Q-Sepharose FF col-umn, and the His6 tag was removed by proteolysis with IIa asoutlined previously (38). His6 tag free protein was separatedfrom the His6 tag and IIa using Superdex 200 gel filtrationchromatography equilibrated with 50 mM Tris-HCl, pH 7.5,containing 100 mM NaCl (TBS). Each batch of purified pro-tein was concentrated to �3 mg/ml using an Amicon centrif-ugal filter device with a 3000Mr cutoff membrane.Protease Inhibition Assay—All reactions were carried out in

TBS, pH 7.5 (50 mM Tris-HCl, pH 7.5, containing 100 mM

NaCl), containing 0.1 mg of BSA/ml (TBS/BSA) and 2 mM

Ca2� (TBS/BSA/Ca2�, pH 7.5). Each enzyme (Pm, pKLK,FXIa, APC, IIa, IIa/sTm, FVIIa/sTF, and tPA) was incubatedwith various concentrations (10�1 to 105 nM) of BPTI (aproti-nin), KD1-WT, or KD1-L17R for 1 h at room temperature ina 96-well microtitration plate (total volume 100 �l). Syntheticsubstrate (5 �l) appropriate for each enzyme was then addedto a final concentration of 1 Km, and residual amidolytic activ-ity was measured in a Vmax kinetic microplate reader (Molec-ular Devices). The inhibition constant, Ki

��, was determinedusing the nonlinear regression data analysis program Grafit.Data for BPTI, KD1-WT, and KD1-L17R were analyzed withan equation for a tight-binding inhibitor (Equation 1) where viand v0 are the inhibited and uninhibited rates, respectively,and [I]0 and [E]0 are the total concentrations of inhibitor andenzyme, respectively (39, 40).

� i � �0

��K*i �� �I0 � �E02 � 4�I0�E0

1⁄2 � �K*i �� �I0 � �E0

2�E0

(Eq. 1)

Inhibition data for APC, IIa, IIa/sTM, tPA, or FVIIa/sTF,where Ki

�� �� [E]0 were analyzed using Equation 2.

� i � �0/�1 � �I0/K*i � (Eq. 2)

Ki� values were obtained by correcting for the effect of sub-

strate according to Beith (39), using Equation 3, where [S] issubstrate concentration and Km is specific for each enzyme.

K*i � K*i �/�1 � �S/KM (Eq. 3)

Coagulation Assays—The inhibitory effects of aprotinin,KD1-WT, and KD1-L17R and TE were measured in the acti-

vated partial thromboplastin time (aPTT) and in dilutethromboplastin time using recombinant TF (24 pM functionalconcentration) prepared as described previously (35). In theaPTT assay, 50 �l of pooled normal human plasma (NPP) wasincubated with 50 �l of aPTT reagent (Diapharma) and 50 �lof test sample in TBS/BSA, pH 7.5, for 5 min at 37 °C. A 50-�lvolume of 35 mM CaCl2 at 37 °C was then added, and the clot-ting time recorded. In the TF-induced clotting assay, 80 �l ofNPP each containing various concentrations of the test com-pound was incubated with 80 �l of dilute recombinant TF for3 min at 37 °C at which point 80 �l of 25 mM CaCl2 at 37 °Cwas added and the clotting time recorded.Fibrinolysis (Clot Lysis) Assay—In principal, the method

followed was of that of Sperzel and Huetter (41) with a fewmodifications. As BPTI (aprotinin) inhibits intrinsic coagula-tion and KD1-WT inhibits both the extrinsic and the intrinsicpathways of coagulation, we chose IIa to initiate clotting andfibrin formation in NPP. The lysis of the formed clot (fibrinol-ysis) was induced by simultaneous addition of tPA. Clot for-mation and lysis were monitored with a microplate reader(SPECTRAmax 190, Molecular Devices) measuring the opti-cal density at 405 nm. Briefly, 10 �l of each test compound orsaline control was added to 240 �l of NPP. Two hundredtwenty five �l of this mixture was then added to 25 �l of IIaand tPA in TBS/BSA containing 25 mM CaCl2. In the 250-�lfinal volume, concentration of IIa was 0.15 �g/ml and that oftPA was 1 �g/ml. Under control conditions (zero tPA andzero test compound), OD405 increased immediately indicatingclotting followed by an extremely slow decrease representingfibrinolysis. As clotting was almost complete after 5 min, fi-brinolysis induced by tPA was evaluated as a relative decreaseof OD405 within 5–30 min. BPTI (aprotinin) was tested at fi-nal concentrations ranging from 0.1 to 8 �M, KD1-WT from0.1 to 60 �M, KD1-L17R from 0.1 to 20 �M, and TE from 0.3�M to 1 mM. Each compound was tested in triplicate.Amino Acid Sequence Analysis and Mass Spectrometry—

Automated N-terminal protein sequencing was performedusing a pulsed-liquid solid phase sequenator with on-line phe-nylthiohydantoin-amino acid analysis (Applied BiosystemscLC system) at the UCLA Biopolymer Core Laboratory di-rected by Dr. Dave Teplow. MALDI-TOF (matrix-assistedlaser desorption/ionization-time of flight) mass spectra (MS)were acquired using Applied Biosystems Voyager-DE-STR atthe UCLAMolecular Instrumentation Center Core Facilitydirected by Dr. Joseph Loo.Molecular Modeling and the Selected Serine Proteases P2�

Residue Specificity—The crystal structures of BPTI (42) andthat of the KD1 (38) with trypsin have been determined. Thecrystal structure of the protease domain of Pm (43) and thatof pKLK (44) have also been determined. Using these struc-tures as templates, we modeled the complexes of proteasedomain of Pm and of pKLK with KD1 with high degree ofaccuracy. The structure of the protease domain of FXIa com-plexed with A�PP (45) was used to obtain the model of FXIa-KD1 complex, and the structure of FVIIa/sTF with a BPTImutant was used to obtain the model of FVIIa-KD1 complex(46). Mutations in KD1 at position 17 were introduced usingthe program O (47). The relative positions of the inhibitors

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4331

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 4: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

and the protease domains of various enzymes were main-tained, and minor adjustments were only made in the sidechains. Hydrophobic/van der Waals, hydrogen bonds, andionic interactions were observed between each protease-in-hibitor complex. All of these interactions were taken into con-sideration in evaluating each inhibitor-protease complex, andit was assumed that all potential hydrogen bond donors andacceptors would participate in these interactions. The proto-cols for modeling these complexes have been described earlier(21). For P2�/S2� subsite specificity, the sequences of the vari-ous coagulation factors and selected other serine proteases aswell as the sequence of Pm from different species were ob-tained from the Uniprot database (48). The ClustalW pro-gram was used for multiple sequence alignment (49).Mouse Liver Laceration Model—A protocol using this

mouse model to study efficacy of KD1 variants was approvedby Chancellor’s Animal Research Committee, UCLA. C57/BL6 mice (8–10 weeks old), weighing �20 g in weight werepurchased from Taconic Farms (Germantown, NY). Animalswere fed standard rodent chow and water and housed in avivarium room with normal room temperature and 12-hlight-dark cycles. Dosage of KD1 variants was comparablewith that used in humans, i.e. 4 �g/g adjusted for mouseweight. A calculated blood level achieved by this dose was �8�g/ml (�1 �M) for each KD1 variant. Animals injected withBPTI (aprotinin, 4 �g/g) or TE (10 �g/g, calculated bloodlevel of 20 �g/ml or �125 �M) were used as positive controls,and those receiving saline were used as negative controls. Allfour antifibrinolytic agents were free of lipopolysaccharide astested using the Limulus amebocyte lysates kit (BioWhittakerInc., Walkersville, MD).Animals were injected intravenously via the tail vein, with

the drug in a volume of 100 �l of sterile balanced salt solutionimmediately preceding anesthesia. Anesthesia was inducedand maintained with Isofluorane. The animals were placed ona heating pad in supine position throughout the procedure tomaintain rectal temperature at 37 °C. The abdomen wasshaved and prepped. Liver was exposed via an anterior rightsubcostal incision. The left lobe of the liver was exteriorizedby gentle continuous pressure on the left flank posteriorly. A5-mm transverse incision was made on the left lobe at 1 cmfrom its inferior edge, using a sharp sterile number 10 scalpelblade. All blood oozing from the site of the left lobe incisionwas collected in a sterile preweighed 3 � 3-cm plastic tray fora total duration of 30 min. Animals were euthanized at theend of the 30-min period. Total blood loss was compared be-tween different groups.Statistical Methods—We compared mean total blood loss

in milligrams from the mouse liver across five treatmentgroups (saline, BPTI, KD1-WT, TE, and KD1-L17R) usingfactorial one-way analysis of variance methods. The p valuefor comparing any two means was computed using a post hoctest under this analysis of variance model. Examination of themean total blood loss error distribution pooled across all fivetreatments showed that it was normal (Gaussian) after con-trolling for the effect of treatment. The Shapiro-Wilk good-ness of fit test was used to formally compare this distribution

to a Gaussian. Thus, no transformations of total blood losswere needed.Toxicity Studies—Toxicity studies were performed by an

intravenous administration of BPTI using 2.5 times and theKD1-L17R using 3 times the dose employed in efficacy experi-ments. Following the injection of a single dose, animals (threein each group) were monitored closely for any obviouschanges in appetite, behavior, activity, limb weakness, andbody weight. The choice of time at which animals were eutha-nized for evaluation of organ injury was based on a time lineof progression of aprotinin-induced renal injury in humans.In BPTI-treated animals, noticeable renal epithelial injury andtubular necrosis were detected on day 10, with a significantresolution by day 21. Accordingly, in a separate experiment,two animals each in BPTI and KD1-L17R group were eutha-nized on days 2, 10, and 20, and a terminal cardiac puncturewas performed to obtain blood samples for renal and liverfunction tests prior to performing autopsies. Organ weight,gross microscopic examination, and full histopathologic ex-amination were also performed.

RESULTS

As a rational approach for mutagenesis of KD1-WT forgain of function in Pm inhibition, we examined the modeledstructures (see “Discussion”) of KD1-WT and of BPTI incomplex with the catalytic domain of human Pm. From thesemodeled structures, it was inferred that changing Leu-17(BPTI numbering) in KD1-WT to Arg would improve Pminhibition and weaken inhibition of the coagulation en-zyme(s). This concept was tested by expression of KD1-L17Rand investigations of its inhibitory properties.Sequence and Mass Spectrometry Analysis of Purified

KD1-WT and KD1-L17R—His6-tagged KD1-WT and KD1-L17R were expressed in E. coli strain BL21(DE3)pLysS. Thefusion proteins were refolded and purified using Q-SepharoseFF column. The His6 tag was then removed by proteolysiswith IIa, and His6 tag-free proteins were separated from theHis6 tag and IIa using Superdex 200 gel filtration chromatog-raphy. The N-terminal sequence of KD1-WT and that ofKD1-L17R was the same (Gly-Ser-His-Met-Asp-Ala-Ala-Gln-Glu-Pro). Clearly, the His tag has been removed, and the pro-teins have the correct sequences. The mass spectra ofKD1-WT and KD1-L17R are given in Fig. 1. The mass of eachprotein is within �1 Da of the expected mass. From the massspectra, we envision that all six Cys residues in the KD1Kunitz domains are involved in making the correct three di-sulfide bonds. This was also seen in the crystal structure ofKD1-WT with trypsin (38). Furthermore, each KD1 prepara-tion migrated as a single band with an apparent molecularmass of �11,000 Da (Fig. 1, inset) in denaturing reduced SDS-polyacrylamide gels (50).Effect of KD1-WT, KD1-L17R, BPTI, and TE on the Clotting

Activity of Normal Plasma—KD1-WT prolonged the clottingtimes of normal plasma both in the aPTT (Fig. 2A) and in thedilute thromboplastin assay (Fig. 2B). BPTI (aprotinin) pro-longed the clotting time of normal plasma only in the aPTTassay (Fig. 2A); however, it was somewhat more potent thanKD1-WT. In contrast, KD1-L17R or TE did not prolong the

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

4332 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 6 • FEBRUARY 11, 2011

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 5: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

normal plasma clot times in either of these two assays. Theseclotting data are consistent with the previous observationswith BPTI (25), TE (41), and full-length TFPI-2 (22). Fromthese observations, we infer that KD1-L17R might not inhibitpKLK, FXIa, or FVIIa/TF. Experimental results presented be-low support this hypothesis.Comparison of the Inhibitory Properties of KD1-WT, KD1-

L17R, and BPTI—In agreement with previous studies (22, 23),KD1-WT inhibited pKLK, FXIa, and Pm with Ki

� values of 25,18, and 6 nM, respectively (Fig. 3 and Table 1). BPTI inhibitedPm with 0.5 nM Ki

� and pKLK with 18 nM Ki�; however, it in-

hibited FXIa weakly with a Ki� value of �346 nM (Fig. 3 and

Table 1). The Ki� values studied here for BPTI for each en-

zyme are in agreement with the values studied earlier(30–32).KD1-L17R did not inhibit pKLK or FXIa; however, it inhib-

ited Pm with high affinity (Ki� �0.9 nM) very similar to BPTI

(Fig. 3 and Table 1). Neither BPTI nor KD1-L17R inhibitedFVIIa/sTF; however, as previously studied (22, 23), KD1-WTinhibited FVIIa/TF with �1 �M Ki

�. Moreover, similar toKD1-WT, the KD1-L17R did not inhibit IIa, IIa/sTM, APC, ortPA (Table 1). In this context, BPTI only inhibited IIa/sTM

and APC with Ki� values of 2–3 �M (Table 1). Collectively, our

data presented in Figs. 2 and 3 corroborate that KD1-L17R isa very specific and potent inhibitor of Pm.In the normal aPTT assay system, higher concentrations of

KD1-WT were needed to inhibit clotting than those predicted

FIGURE 1. MALDI-TOF mass spectra and SDS-PAGE analysis of KD1-WTand KD1-L17R. The mass spectra of KD1-WT (top) and KD1-L17R (bottom)obtained using Applied Biosystems Voyager-DE-STR are depicted. The spec-tra were calibrated so that the masses should be within �1 Da. KD1-WTmeasured was 8844.8 Da and predicted was 8843.9 Da; KD1 L17R measuredwas 8888.2 Da and predicted was 8886.9 Da. Inset, reduced SDS-PAGE (15%acrylamide) of KD1-WT (top) and of KD1-L17R (bottom). Lane 1, mixture ofreduced standard proteins; lane 2, 5 �g of KD1-WT (top) or 5 �g of KD1-L17R (bottom).

FIGURE 2. Inhibition of plasma coagulation by BPTI, KD1-WT, KD1-L17R,and TE. A, activated partial thromboplastin time (aPTT); B, dilute thrombo-plastin time (dilute PT). Clotting times were obtained as outlined under “Ex-perimental Procedures.”

FIGURE 3. Determination of equilibrium dissociation constants (Ki�) of

KD1-WT, KD1-L17R, and BPTI with pKLK, FXIa, and Pm. The enzyme ac-tivity is expressed as the percent fractional activity (inhibited rate/uninhib-ited rate) at increasing inhibitor concentrations. The dissociation constants(Ki

�) were determined using Equations 1 and 2 as outlined under “Experi-mental Procedures.” Shown in A is the percent activity of 1 nM pKLK remain-ing in the presence of various concentrations of KD1-WT (E), KD1-L17R (F),and BPTI (�). Shown in B is the percent activity of 1 nM FXIa remaining in thepresence of various concentrations of KD1-WT (E), KD1-L17R (F), and BPTI(�). Shown in C is the percent activity of 3 nM Pm remaining in the presenceof various concentrations of KD1-WT (E), KD1-L17R (F), and BPTI (�). The Ki

values are listed in Table 1.

TABLE 1Ki

� values for the inhibition of selected coagulation proteases, tPAand Pm, by KD1-WT, KD1-L17R, and BPTI

EnzymeKi

� valuesa

KD1-WT KD1-L17R BPTI

nMpKLK 25 � 3 �3000b 18 � 2FXIa 18 � 2 �3000b 346 � 25Pm 6 � 0.5 0.9 � 0.1 0.5 � 0.1VIIa/sTF �1000 �3000b �3000bIIa �3000b �3000b �3000bIIa/sTM �3000b �3000b �3000APC �3000b �3000b �2000tPA �3000b �3000b �3000b

a All Ki� values represent an average of at least three independent measurements.

b Data represent lower limit Ki� values; the actual values could not be determined.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4333

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 6: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

from the Ki� value for FXIa inhibition. A most likely explana-

tion for this could be that aPTT is a kinetic based assay,whereas the equilibrium inhibition constant Ki

� was obtainedupon prolonged incubation (1 h) of the inhibitor KD1-WTwith the enzyme FXIa.Fibrinolysis Assay—The objective of these experiments was

to evaluate the potency of KD1-L17R in inhibiting tPA-in-duced plasma clot fibrinolysis. As shown in Fig. 4A, additionof IIa to NPP results in an increase in OD405 reflective of astable fibrin clot formation (Œ); however, simultaneous addi-tion of tPA results in a biphasic curve with an initial increasein OD405 (fibrin formation), which is followed by a rapid de-crease associated with the fibrinolytic activity (Fig. 4A, �) in-duced by Pm that is formed by the action of tPA on plasmino-gen. Using KD1-L17R as a representation, it reveals thatantifibrinolytic agents inhibit fibrinolysis in a dose-dependentmanner under these conditions (Fig. 4A). Such data were col-lected with each antifibrinolytic compound (BPTI, KD1-L17R,Kd1-WT, and TE). To better represent these experiments,results are presented as percent clot lysed at 12 min (the timewhen fibrin clot is completely lysed in the absence of inhibi-tor) as a function of inhibitor concentration (Fig. 4B). Under

these conditions, BPTI inhibited fibrinolysis with an apparentIC50 of 0.31 � 0.04 �M, KD1-L17R with an apparent IC50 of0.36 � 0.05 �M, KD1-WT with an apparent IC50 of 1.52 �0.24 �M, and TE with an apparent IC50 of 15.2 � 1.71 �M,respectively. These results are in good agreement with theantifibrinolytic activity of BPTI and TE obtained by Sperzeland Huetter (41) under similar conditions. Of significance isthe observation that KD1-L17R is very effective in inhibitingplasma clot fibrinolysis that is quite comparable with BPTI.Mouse Liver Laceration Model—Because tPA and excessive

fibrinolysis is the main cause of bleeding in the liver lacerationanimal model (51), we evaluated the effects of BPTI, KD1-WT, KD1-L17R, and TE on blood loss in this animal model.These in vivo experiments complement the in vitro studiespresented in Fig. 4. The effects of all four antifibrinolyticagents on the amount of blood loss are depicted in Fig. 5. Ascompared with saline, the amount of blood loss was reducedby �84% by KD1-L17R (p 0.0005), by �70% by BPTI (p0.0036), �52% by TE (p 0.0196), and only �10% by KD1-WT(p 0.6505). Clearly, KD1-L17R is very effective in reducingblood loss in this animal model. Statistically, there was nosignificant difference in the amount of blood loss betweenKD1-L17R and BPTI-treated animals (p 0.5223). Further-more, at this point, we cannot conclude whether there is asignificant difference in the amount of blood loss betweenKD1-L17R and TE-treated animals (p 0.1312). Additionalexperiments using �40 animals in each group would berequired to establish whether or not KD1-L17R is superiorto BPTI and/or TE. In contrast, KD1-L17R was significantlyeffective compared with the KD1-WT treated animals (p0.0017).Toxicity Studies—Out of the eight animals that were given

an intravenous dose of BPTI (2.5� the efficacy dose) or KD1-L17R (3� the efficacy dose), necropsy was performed on twoanimals in each group on the following days, 2, 10, and 20. Inthe BPTI group, no injury was noted on day 2; moderate de-

FIGURE 4. Effect of KD1-WT, KD1-L17R, BPTI, and TE on fibrinolysis inhuman NPP. A, evaluation of fibrinolysis in NPP. IIa was added to NPP toinitiate fibrin clot formation, which is associated with an increase in opticaldensity at 405 nm (OD405). The clot was stable for 4 h; however, for clarity,the data are shown for up to 30 min only (Œ). Simultaneously added tPAconverted plasminogen to Pm, which dissolved the fibrin clot completelywithin �12 min, as indicated by an initial increase followed by a decrease inOD405 (�). Addition of KD1-L17R inhibited fibrinolysis in a dose-dependentmanner as follows: 0.5 �M (F), 1 �M (E), 2 �M (f), and 4 �M (�), respec-tively. For comparison, such data were collected for all four antifibrinolyticcompounds at varying concentrations, and fibrinolysis was evaluated as the% lysis (decrease in OD405) at 12 min, the time at which the fibrin clot iscompletely lysed without the added inhibitor. B, evaluation of the potencyof various antifibrinolytic agents. BPTI was tested at concentrations rangingfrom 0.01 to 8 �M, KD1-L17R from 0.1 to 20 �M, KD1-WT from 0.1 to 60 �M,and TE from 0.3 to 1000 �M. The data are presented as average of three de-terminations. Percent clot lysis (y axis) at 12 min is plotted against inhibitorconcentration (x axis, log scale). BPTI, �; KD1-L17R, F; KD1-WT, E; and TE, Œ.

FIGURE 5. Effect of KD1-WT, KD1-L17R, BPTI, and TE in a mouse liverlaceration bleeding model. Animals were treated with saline, KD1-WT,KD1-L17R, BPTI, or TE by bolus injection, and - mm transverse incision wasmade on the left lobe at 1 cm from its inferior edge, using a sharp sterilenumber 10 scalpel blade. All blood oozing from the site of the left lobe inci-sion was collected in a sterile preweighed 3 � 3-cm plastic tray for a totalduration of 30 min. Total blood loss was compared between differentgroups. Data are presented as mean � S.E. of 10 animals. Probability p val-ues between different groups were as follows: 1) saline versus KD1-L17R,0.0005; saline versus BPTI, 0.0036; saline versus TE, 0.0196; saline versus Kd1-WT, 0.6505; 2) KD1-L17R versus BPTI, 0.5223; KD1-L17R versus TE, 0.1312;KD1-L17R versus KD1-WT, 0.0017; and 3) KD1-WT versus BPTI, 0.0118;KD1-WT versus TE, 0.0578. *, p value 0.01 compared with saline control.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

4334 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 6 • FEBRUARY 11, 2011

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 7: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

gree of multifocal renal tubular epithelial necrosis was notedon day 10 (Fig. 6A); and scattered areas of tubular necrosiswith many areas of regeneration was noted on day 20. No in-jury was noted in KD1-L17R group (Fig. 6B) on any of thesedays. However, renal function tests such as blood urea nitro-gen and serum creatinine were normal and remained essen-tially unchanged in both groups. Our animal data on BPTItoxicity corroborate previous findings that aprotinin causesrenal dysfunction in humans (27, 52).

DISCUSSION

Kunitz-type domains are compact stable modules and havebeen widely used for therapeutics where the extent of proteol-ysis must be carefully controlled; however, they lack specific-

ity and often inhibit the target protease as well as anti-targetproteases. For example, KD1 of TFPI-2 inhibits intrinsic andextrinsic coagulation as well as Pm, a fibrinolytic enzyme. Theobjective of this study was to minimally mutate KD1 such thatthe mutant molecule strongly inhibits plasmin without inhib-iting extrinsic and/or the intrinsic pathway of coagulation. Toaccomplish this goal, we modeled complexes of protease do-main of Pm and of selected coagulation proteases with KD1.We examined S2�/P2� subsite interactions in these proteaseinhibitor complexes and predicted that changing residueLeu-17 to Arg in KD1 might abrogate the anticoagulant prop-erty of KD1 while simultaneously enhancing Pm inhibition.Experimental evidence presented in Figs. 2 and 3 supportsthis contention. The molecular models of protease domain ofPm with KD1-WT and mutants as well as those of pKLK,FXIa, and FVIIa with KD1-WT and KD1-L17R discussed be-low provide structural support for the experimental results.Pm Protease Domain-KD1 Structural Model and S2� Sub-

site Specificity—The protease-inhibitor complexes of Pm withKD1-WT and the mutants are shown in Fig. 7. Clearly the S2�subsite pocket in Pm is outlined by strong electronegativesurface that is primarily contributed by the side chain of resi-dues Glu-73 and Glu-143 (chymotrypsin numbering). Al-though a small hydrophobic residues such as Leu can be ac-commodated in this pocket (Fig. 7A), a basic residue such asArg is preferentially suited to interact with Glu-73 of Pm (Fig.7B). In agreement with this concept, a search for the substratepreference for Pm reveals that it prefers P2� Arg, Lys, Ser, orThr residue at the S2� site (Table 2). Similar to Arg at position17 in KD1, Lys can also make an H-bond with Glu-73 of Pm(Fig. 7C). Because Arg is much more basic than Lys, it is ex-pected that Arg will make stronger interactions with Glu-73of Pm and contribute toward higher affinity (32).Although Glu-143 contributes to S2� electronegative poten-

tial, its side chain conformation along with that of Arg/Lys 17could not be adjusted to make H-bond interactions. Withoutadjusting the main chain conformation, a constraint imposedon modeling, major steric conflicts were encountered for in-teraction of Arg/Lys-17 with Glu-143 of Pm to occur. In con-trast, residue Thr (or Ser) at position 17 in KD1 could makean H-bond with Glu-143 directly (Fig. 7E) or through a watermolecule with Glu-73 (Fig. 7F) in Pm. Further work, especiallyx-ray crystal structures, is needed to precisely define the S2�/P2� interactions between Pm with KD1-WT and the P2� siteKD1 mutants.It has been shown that Pm cleaves FVIII and FVa at sites

where the P2� residue is Gln (Table 2) (59, 65). Although Glnat P2� position 17 in KD1 could be fitted into the S2� pocketand might interact with Glu-73 of Pm through a water mole-cule (Fig. 7D), this interaction is not favorable due to two rea-sons. First, Gln side chain is neutral but rich in lone electronpairs, and the S2� pocket itself is electronegative and rich inelectrons. Second, interaction of neutral Gln (as comparedwith basic Arg or Lys) with Glu-73 of Pm is expected to beweaker. These considerations are consistent with previousobservations that KD1-L17Q is a weak inhibitor of Pm (23).

FIGURE 6. Renal tubular necrosis with BPTI but not KD1-L17R. Photomi-crographs of kidneys from mice 10 days after the injection of BPTI (A) andKD1-L17R (B) are shown. The dosage used was 2.5� for BPTI and 3� forKD1-L17R of the dose used in efficacy studies (Fig. 5). Tissues were fixed informalin and stained with hematoxylin and eosin. Magnification is 40�.Thin arrow depicts tubular necrosis, and thick arrow depicts normal tubularepithelium in A. Tubular necrosis was not seen in group B.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4335

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 8: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

pKLK-KD1, FXIa-KD1, and FVIIa-KD1 Structural Modelsand S2� Subsite Specificity—The protease inhibitor complexesof pKLK, FXIa, and FVIIa with KD1-WT and KD1-L17R aregiven in supplemental Fig. S1. The S2� subsite in these coagu-lation proteases consists of a hydrophobic pocket that prefersto accommodate a nonpolar residue such as P2� Leu-17. Thus,

the Arg or Lys residue at the P2� position would not be favor-able. In support of this, a search for substrate specificity ofcoagulation proteases, including pKLK, FXIa, FVIIa, as well astPA, uPA, matriptase-2, and APC reveals that none of thesubstrates of these proteases contain Arg/Lys at P2� position(Table 2). In pKLK or FVIIa, residue 73 is Leu, and it is Gln in

FIGURE 7. Modeled complexes of the protease domain of Pm with KD1-WT and Leu-17 mutants. Ribbon diagram of the overall structures of the com-plex of KD1-WT (yellow) with the protease domain of Pm (red) is shown on the left. The Leu-17 (BPTI numbering) of KD1-WT is shown in a stick representa-tion. Enlarged views of the PyMol (53)-generated maps of the electrostatic potential at the solvent-accessible surface of the plasmin catalytic domain nearresidue 17 of KD1 are shown on the right. The residues within 6 Å radius of Leu-17 of KD1 were selected for display. The wild-type residue Leu-17 in KD1 isshown in A. The effect of mutation of KD1 Leu-17 to Arg is shown in B, to Lys in C, to Gln in D, and to Thr in E and F. Blue represents positive, red representsnegative, and white represents hydrophobic surface potential. Red sphere represents water molecule. Single letter code and chymotrypsin numbering isused for residues of the protease domain of Pm as follows: M39 {585}, H40 {586}, F41 {587}, C42 {588}, E73 {623}, W141 {685}, G142 {686}, E143 {687}, Q192{738}, G193 {739}, and D194 {740}. The numbers in braces represent Pm numbering. H-bonds are shown by dashed lines.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

4336 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 6 • FEBRUARY 11, 2011

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 9: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

FXIa. Although Gln-73 in FXIa has the potential to interactwith Arg-17 in KD1, the interaction would be weaker as com-pared with Glu-73 in Pm. Moreover, conformation of the sidechain of Gln-73 in FXIa is stabilized by two H-bonds, onewith His-34 through its carbonyl oxygen, and the second withThr-32 through its NH2 group (45).Our data presented in Figs. 2 and 3 are consistent with the

concept that coagulation proteases prefer a nonpolar residueat P2� position in their substrates/inhibitors (6, 17, 23, 42). Inthis context, KD1-L17R has no inhibitory activity towardpKLK, FXIa, and FVIIa/sTF. This concept is also supported bythe inhibitory profile of BPTI, which has P2� Arg residue atposition 17 (42). BPTI is a very strong inhibitor of Pm (Ki

�0.5 nM) as compared with pKLK (Ki� �20 nM), FXIa (Ki

�350 nM), or FVIIa/sTF (Ki� �3 �M). In support of this, mod-

eling of protease domain of Pm in complex with BPTI re-vealed that Arg-17 of BPTI makes a salt bridge with Glu-73 ofPm (data not shown).Nature of Residues 73 and 143 in Pm and Other Serine

Proteases—The primary residues that provide negative elec-trostatic potential in S2� pocket of Pm are residues Glu-73and Glu-143. A data base search reveals that these residuesare conserved in Pm from all species (Table 3). Furthermore,instead of an acidic residue, all coagulation proteases have

neutral or basic residues at positions 73 and 143 (Table 3).Thus, it would appear that coagulation proteases prefer a hy-drophobic residue at the P2� position in their substrates,whereas Pm prefers a basic residue at this position. Therefore,coagulation proteases and Pm have different substrate speci-ficities at P2� position and might be used for development ofselective synthetic inhibitors and/or substrates.Pharmacologic Implications—First, the choice of the liver

laceration model was based on documented enhancement offibrinolysis in the setting of liver trauma in the clinical situa-tion (51). This mimics the clinical indications for which BPTI(aprotinin) was used in the setting of enhanced fibrinolysissuch as cardiac surgery, extremity, and organ trauma and or-gan transplant. Second, from the enzyme inhibition data andthe in vivo animal data, KD1-L17R proved to be a very potentinhibitor of tPA-induced fibrin degradation and blood loss.The dose of KD1-L17R used was the same as the dose forBPTI (aprotinin) in clinical use. Based on calculated totalbody water, the dose of either BPTI or KD1-L17R translatesto a plasma level of �8 �g/ml. This concentration is wellabove that required for effective inhibition in vitro of Pm byboth BPTI and KD1-L17R (Figs. 3 and 4). Moreover, BPTI hasa half-life of 2.5 h. KD1-L17R is similar in size to BPTI, andalthough no pharmacokinetic studies have been performed, it

TABLE 2Specificity of P2� subsite of Pm versus selected human coagulation serine proteases, matriptase and tPA/urokinase-typeAmino acid sequences of the substrates from P3 to P3� subsites are shown. The arrow depicts the cleavage site in each case. P2� residue is in boldface. The listedsequences cleaved by Pm are those that have not been reported to be cleaved by the other serine proteases. Note that Pm prefers Arg, Lys, Ser, or Thr at the P2� subsitein its substrates/inhibitors, whereas the other proteases prefer nonpolar residues at this subsite. The abbreviations used are as follows: HMWK, high molecular weightkininogen; Fgn, fibrinogen; PAR, protease-activated receptor; Pgn, plasminogen; uPAR, urokinase plasminogen activator receptor; VEGF, vascular endothelial growthfactor; CgA, chromogranin; OPN, osteopontin; MMP3, matrix metalloprotease 3; EPI, extrinsic pathway inhibitor same as TFPI type-1; uPA, urokinase Pgn activator.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4337

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 10: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

is presumed that its half-life would be about the same asBPTI. Therefore, a single dose given in an animal modelwhere the total duration of bleeding is no more than 30 min ispresumed to generate sufficient levels for the length of theexperiment. Under these conditions, KD1-L17R was as effec-tive or better in preventing blood loss either by BPTI or TE(Fig. 5). Although we did not challenge the animals hemo-statically with antiplatelet agents or anticoagulants, we specu-late that under such circumstances the dose may be repeatedand/or the animal given a continuous infusion of the drug forenhancing its antifibrinolytic efficacy. Studies of the efficacyof KD1-L17R as an antifibrinolytic agent in hemostaticallychallenged animals and its pharmacokinetic studies areplanned.KD1-L17R does not induce renal toxicity in mice when

used at higher doses (3�) compared with BPTI (2.5�). Anargument may be made that toxicity studies were performedin normal animals not challenged by the perturbations of pre-existing medical conditions, nephrotoxic medications, pro-longed general anesthesia, and blood loss, all of which predis-pose the organism to renal damage, as is the case clinicallywhere BPTI was used. Nonetheless, BPTI used under similarconditions did cause obvious histopathologic changes as seenin Fig. 6. A plausible explanation for why there was no eleva-tion in indices of renal function such as blood urea nitrogenand serum creatinine despite histopathologic changes, in ani-mals administered toxic doses of BPTI, is that a considerable

loss of nephron function is required before renal functiontests become elevated. Therefore, it is conceivable that in asetting with above predisposing factors, BPTI could easilylead to detectable abnormality in renal function tests.Two possible explanations for why KD1-L17R may not

cause renal damage are as follows: 1) its acidic charge (isoelec-tric point 5.0, net charge �2.8, pH 7.5), which precludes itfrom collecting in the renal tubular epithelium as has beendescribed with BPTI (isoelectric point 8.95, net charge �5.2,pH 7.5); 2) the finding that KD1-L17R does not inhibit pKLK,which leads to altered microcirculation in kidneys. Further-more, KD1-L17R did not cause any detectable histopathologicchanges in any other major organ, including heart, lungs,brain, and liver. KD1-L17R (and BPTI) also did not cause anyseizures as indicated for TE (71, 72).Conclusions—From the in vitro antifibrinolytic data and the

in vivo animal data presented here, it is clear that KD1-L17Ris a very potent inhibitor of tPA-induced fibrin degradationand loss of blood. KD1-L17R is also a specific inhibitor of Pmand does not inhibit pKLK, FXIa, or FVIIa/TF as opposed tothe native TFPI-2 or KD1-WT. In this context, it should benoted that TFPI-2 has been proposed to be used as an antico-agulant (73); however, its anti-Pm activity could offset its pro-posed beneficial effect. Similarly, KD1-WT has been proposedto be used as anti-Pm agent (74); however, its anticoagulantactivity could offset such an effect. Moreover, data presentedin this study do not support any efficacy of KD1-WT in the

TABLE 3Amino acid sequence alignment of 70 – 80 and 140 –155 loops (chymotrypsin numbering) of Pm and selected serine proteases surroundingthe S2� subsite pocketResidues 73 and 143 are shaded and are acidic in Pm as compared with the other serine proteases involved in blood coagulation. To aid the reader in identifying the 70–80 and 140–150 segment positions in each protease, the actual sequence numbers (where available) are also given in braces following the name of the protease.

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

4338 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 6 • FEBRUARY 11, 2011

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 11: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

liver laceration mouse injury model. Furthermore, KD1-R15K(BPTI numbering) has also been proposed to be used as ananti-Pm agent (74); however, no supporting evidence is pre-sented as to its efficacy in any animal model.KD1-L17R does not induce renal toxicity as compared with

BPTI or seizures as is the case with TE. Moreover, it is of hu-man origin and differs only from the native protein by oneamino acid as compared with EPI-302 (32), which differs inseven residues from the native TFPI Kunitz domain. Thus,KD1-L17R might not be immunogenic. Furthermore, EPI-K503/DX-88/Ecallantide, a specific inhibitor of pKLK, wasnot effective in reducing blood loss in on-pump cardiotho-racic surgery (75), where BPTI is very effective in this setting.Thus, the effect of BPTI is due to its ability to inhibit Pm andnot pKLK. KD1-L17R inhibits Pm and none of the coagula-tion enzymes, including pKLK. Thus, it could represent as analternative to BPTI (aprotinin) therapy in clinical settingswhere excessive blood loss is anticipated. Importantly, ourdata indicate that S2�/P2� subsite interactions play a majorrole in Pm/inhibitor selectivity.

Acknowledgments—We thank Dr. Jeffery Gornbein of Department ofBiomathematics, UCLA, for help with the statistical analysis of theanimal bleeding data. We also thank Dr. Teplow for amino acidsequence analysis and Dr. Loo for MALDI-TOF MS analysis.

REFERENCES1. Neurath, H., and Walsh, K. A. (1976) Proc. Natl. Acad. Sci. U.S.A. 73,

3825–38322. Huber, R., and Bode, W. (1978) Acc. Chem. Res. 11, 114–1223. Whitcomb, D. C., and Lowe, M. E. (2007) Dig. Dis. Sci. 52, 1–174. Davie, E. W. (2003) J. Biol. Chem. 278, 50819–508325. Plow, E. F., and Hoover-Plow, J. (2004) Trends Cardiovasc. Med. 14,

180–1866. Debela, M., Magdolen, V., Schechter, N., Valachova, M., Lottspeich, F.,

Craik, C. S., Choe, Y., Bode, W., and Goettig, P. (2006) J. Biol. Chem.281, 25678–25688

7. Gros, P., Milder, F. J., and Janssen, B. J. (2008) Nat. Rev. Immunol. 8,48–58

8. Wu, J., Liu, W., Bemis, A., Wang, E., Qiu, Y., Morris, E. A., Flannery,C. R., and Yang, Z. (2007) Arthritis Rheum. 56, 3675–3684

9. Daci, E., Everts, V., Torrekens, S., Van Herck, E., Tigchelaar-Gutterr, W.,Bouillon, R., and Carmeliet, G. (2003) J. Bone Miner. Res. 18, 1167–1176

10. Ebisch, I. M., Steegers-Theunissen, R. P., Sweep, F. C., Zielhuis, G. A.,Geurts-Moespot, A., and Thomas, C. M. (2007) Fertil. Steril. 87,619–626

11. Kunitz, M., and Northrop, J. H. (1936) J. Gen. Physiol. 19, 991–100712. Kazal, L. A., Spicer, D. S., and Brahinsky, R. A. (1948) J. Am. Chem. Soc.

70, 3034–304013. Potempa, J., Korzus, E., and Travis, J. (1994) J. Biol. Chem. 269,

15957–1596014. Wiedow, O., Schroder, J. M., Gregory, H., Young, J. A., and Christo-

phers, E. (1990) J. Biol. Chem. 265, 14791–1479515. Laskowski, M., Jr., and Kato, I. (1980) Annu. Rev. Biochem. 49, 593–62616. Girard, T. J., Warren, L. A., Novotny, W. F., Likert, K. M., Brown, S. G.,

Miletich, J. P., and Broze, G. J., Jr. (1989) Nature 338, 518–52017. Sprecher, C. A., Kisiel, W., Mathewes, S., and Foster, D. C. (1994) Proc.

Natl. Acad. Sci. U.S.A. 91, 3353–335718. Delaria, K. A., Muller, D. K., Marlor, C. W., Brown, J. E., Das, R. C., Roc-

zniak, S. O., and Tamburini, P. P. (1997) J. Biol. Chem. 272,12209–12214

19. Ponte, P., Gonzalez-DeWhitt, P., Schilling, J., Miller, J., Hsu, D., Green-berg, B., Davis, K., Wallace, W., Lieberburg, I., and Fuller, F. (1988) Na-

ture 331, 525–52720. Petersen, L. C., Bjørn, S. E., Olsen, O. H., Nordfang, O., Norris, F., and

Norris, K. (1996) Eur. J. Biochem. 235, 310–31621. Bajaj, M. S., Birktoft, J. J., Steer, S. A., and Bajaj, S. P. (2001) Thromb.

Haemost. 86, 959–97222. Petersen, L. C., Sprecher, C. A., Foster, D. C., Blumberg, H., Hamamoto,

T., and Kisiel, W. (1996) Biochemistry 35, 266–27223. Chand, H. S., Schmidt, A. E., Bajaj, S. P., and Kisiel, W. (2004) J. Biol.

Chem. 279, 17500–1750724. Ohri, S. K., Parratt, R., White, T., Becket, J., Brannan, J. J., Hunt, B. J.,

and Taylor, K. M. (2001) Perfusion 16, 199–20625. Fritz, H., and Wunderer, G. (1983) Arzneimittel-Forschung Res. 33,

479–49426. Royston, D., van Haaften, N., and De Vooght, P. (2007) Eur. J. Anaesthe-

siol. 24, 6–1427. Mangano, D. T., Tudor, I. C., and Dietzel, C. (2006) N. Engl. J. Med. 354,

353–36528. Immer, F. F., Jent, P., Englberger, L., Stalder, M., Gygax, E., Carrel, T. P.,

and Tevaearai, H. T. (2008) Heart Surg. Forum 11, E9–E1229. Beierlein, W., Scheule, A. M., Dietrich, W., and Ziemer, G. (2005) Ann.

Thorac. Surg. 79, 741–74830. Dennis, M. S., Herzka, A., and Lazarus, R. A. (1995) J. Biol. Chem. 270,

25411–2541731. Markland, W., Ley, A. C., and Ladner, R. C. (1996) Biochemistry 35,

8058–806732. Markland, W., Ley, A. C., Lee, S. W., and Ladner, R. C. (1996) Biochem-

istry 35, 8045–805733. Caballero, T., and Lopez-Serrano, C. (2006) J. Allergy Clin. Immunol.

117, 476–47734. Schechter, I., and Berger, A. (1967) Biochem. Biophys. Res. Commun. 27,

157–16235. Bajaj, S. P., Schmidt, A. E., Agah, S., Bajaj, M. S., and Padmanabhan, K.

(2006) J. Biol. Chem. 281, 24873–2488836. Sambrook, J., and Russel, D. W. (2001)Molecular Cloning: A Laboratory

Manual, 3rd Ed., Cold Spring Harbor Laboratory Press, Cold SpringHarbor, NY

37. Stone, M. J., Ruf, W., Miles, D. J., Edgington, T. S., and Wright, P. E.(1995) Biochem. J. 310, 605–614

38. Schmidt, A. E., Chand, H. S., Cascio, D., Kisiel, W., and Bajaj, S. P. (2005)J. Biol. Chem. 280, 27832–27838

39. Beith, J. G. (1984) Biochem. Med. 32, 287–39740. Morrison, J. F., and Walsh, C. T. (1988) Adv. Enzymol. Relat. Areas Mol.

Biol. 61, 201–30141. Sperzel, M., and Huetter, J. (2007) J. Thromb. Haemost. 10, 2113–211842. Helland, R., Otlewski, J., Sundheim, O., Dadlez, M., and Smalås, A. O.

(1999) J. Mol. Biol. 287, 923–94243. Parry, M. A., Fernandez-Catalan, C., Bergner, A., Huber, R., Hopfner,

K. P., Schlott, B., Guhrs, K. H., and Bode, W. (1998) Nat. Struct. Biol. 5,917–923

44. Tang, J., Yu, C. L., Williams, S. R., Springman, E., Jeffery, D., Sprengeler,P. A., Estevez, A., Sampang, J., Shrader, W., Spencer, J., Young, W.,McGrath, M., and Katz, B. A. (2005) J. Biol. Chem. 280, 41077–41089

45. Navaneetham, D., Jin, L., Pandey, P., Strickler, J. E., Babine, R. E., Abdel-Meguid, S. S., and Walsh, P. N. (2005) J. Biol. Chem. 280, 36165–36175

46. Zhang, E., St Charles, R., and Tulinsky, A. (1999) J. Mol. Biol. 285,2089–2104

47. Jones, T. A., Zou, J. Y., Cowan, S. W., and Kjeldgaard, M. (1991) ActaCrystallogr. A. 47, 110–119

48. Jain, E., Bairoch, A., Duvaud, S., Phan, I., Redaschi, N., Suzek, B. E., Mar-tin, M. J., McGarvey, P., and Gasteiger, E. (2009) BMC Bioinformatics10, 136

49. Larkin, M. A., Blackshields, G., Brown, N. P., Chenna, R., McGettigan,P. A., McWilliam, H., Valentin, F., Wallace, I. M., Wilm, A., Lopez, R.,Thompson, J. D., Gibson, T. J., and Higgins, D. G. (2007) Bioinformatics23, 2947–2948

50. Laemmli, U. K. (1970) Nature 227, 680–68551. Groh, J., Welte, M., Azad, S. C., and Kratzer, M. A. (1993) Infusionsther.

Transfusion Med. 4, 173–179

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

FEBRUARY 11, 2011 • VOLUME 286 • NUMBER 6 JOURNAL OF BIOLOGICAL CHEMISTRY 4339

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 12: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

52. Karkouti, K., Beattie, W. S., Dattilo, K. M., McCluskey, S. A., Ghannam,M., Hamdy, A., Wijeysundera, D. N., Fedorko, L., and Yau, T. M. (2006)Transfusion 46, 327–338

53. The PyMOL Molecular Graphics System (2010) Version 1.3, Schro-dinger, LLC, New York

54. Takagi, T., and Doolittle, R. F. (1975) Biochemistry 14, 940–94655. Samis, J. A., Ramsey, G. D., Walker, J. B., Nesheim, M. E., and Giles,

A. R. (2000) Blood 95, 943–95156. Pryzdial, E. L., Lavigne, N., Dupuis, N., and Kessler, G. E. (1999) J. Biol.

Chem. 274, 8500–850557. Beaufort, N., Leduc, D., Rousselle, J. C., Namane, A., Chignard, M., and

Pidard, D. (2004) FEBS Lett. 574, 89–9458. Jiang, X., and Couchman, J. R. (2003) J. Histochem. Cytochem. 51,

1393–141059. Jiang, Q., Taupenot, L., Mahata, S. K., Mahata, M., O’Connor, D. T.,

Miles, L. A., and Parmer, R. J. (2001) J. Biol. Chem. 276, 25022–2502960. Nogami, K., Shima, M., Matsumoto, T., Nishiya, K., Tanaka, I., and Yo-

shioka, A. (2007) J. Biol. Chem. 282, 5287–529561. Christensen, B., Schack, L., Klaning, E., and Sørensen, E. S. (2010) J. Biol.

Chem. 285, 7929–793762. Nagase, H., Enghild, J. J., Suzuki, K., and Salvesen, G. (1990) Biochemis-

try 29, 5783–578963. McSweeney, P. L. H., Olson, N. F., Fox, P. F., Healy, A., and Højrup, P.

(1993) Food Biotech. 7, 143–15864. Hervio, L. S., Coombs, G. S., Bergstrom, R. C., Trivedi, K., Corey, D. R.,

and Madison, E. L. (2000) Chem. Biol. 7, 443–453

65. Mann, K. G., and Kalafatis, M. (2003) Blood 101, 20–3066. Colman, R. W. (2006) in Hemostasis and Thrombosis: Basic Principles

and Clinical Practice (Colman, R. W., Marder, V. J., Clowes, A. W.,George, J. N., and Goldhaber, S. Z., eds) 5th Ed., pp. 107–130, LippincottWilliams &Wilkins, Philadelphia

67. Davie, E. W., and Greenberg, D. L. (2006) in Hemostasis and Thrombo-sis: Basic Principles and Clinical Practice (Colman, R. W., Marder, V. J.,Clowes, A. W., George, J. N., and Goldhaber, S. Z., eds) 5th Ed., pp.21–58, Lippincott Williams &Wilkins, Philadelphia

68. Eaton, D., Rodriguez, H., and Vehar, G. A. (1986) Biochemistry 25,505–512

69. Macfarlane, S. R., Seatter, M. J., Kanke, T., Hunter, G. D., and Plevin, R.(2001) Pharmacol. Rev. 53, 245–282

70. Booth, N. A., and Bachmann, F. (2006) in Hemostasis and Thrombosis:Basic Principles and Clinical Practice (Colman, R. W., Marder, V. J.,Clowes, A. W., George, J. N., and Goldhaber, S. Z., eds) 5th Ed., pp.365–380, Lippincott Williams &Wilkins, Philadelphia

71. Breuer, T., Martin, K., Wilhelm, M., Wiesner, G., Schreiber, C., Hess, J.,Lange, R., and Tassani, P. (2009) Eur. J. Cardiothorac. Surg. 35, 167–171

72. Murkin, J. M., Falter, F., Granton, J., Young, B., Burt, C., and Chu, M.(2010) Anesth. Analg. 110, 350–353

73. Sprecher, C. A., Kisiel, W., and Foster, D. C. (March 17, 1998) U. S.Patent 5,728,674

74. Kisiel, W., and Chand, H. S. (October 7, 2008) U. S. Patent 7,432,23875. Lehmann, A. (2008) Expert Opin. Biol. Ther. 8, 1187–1199

Human Kunitz-type Variant with Enhanced Plasmin Inhibition

4340 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 286 • NUMBER 6 • FEBRUARY 11, 2011

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from

Page 13: EngineeringKunitzDomain1(KD1)ofHumanTissueFactor ... · Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accord- ingly,

Lawson, Sreejesh Shanker, Amy E. Schmidt and S. Paul BajajMadhu S. Bajaj, Godwin I. Ogueli, Yogesh Kumar, Kanagasabai Vadivel, Gregoryto Selectively Inhibit Fibrinolysis: PROPERTIES OF KD1-L17R VARIANT

Engineering Kunitz Domain 1 (KD1) of Human Tissue Factor Pathway Inhibitor-2

doi: 10.1074/jbc.M110.191163 originally published online November 29, 20102011, 286:4329-4340.J. Biol. Chem. 

  10.1074/jbc.M110.191163Access the most updated version of this article at doi:

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

http://www.jbc.org/content/suppl/2011/01/26/286.6.4329.DC1Read an Author Profile for this article at 

Supplemental material:

  http://www.jbc.org/content/suppl/2010/11/29/M110.191163.DC1

  http://www.jbc.org/content/286/6/4329.full.html#ref-list-1

This article cites 68 references, 22 of which can be accessed free at

by guest on August 20, 2019

http://ww

w.jbc.org/

Dow

nloaded from