functionalcharacterizationofthecomplement ... · tural homolog of complement control proteins named...

11
Functional Characterization of the Complement Control Protein Homolog of Herpesvirus Saimiri ARG-118 IS CRITICAL FOR FACTOR I COFACTOR ACTIVITIES * S Received for publication, March 31, 2006, and in revised form, June 7, 2006 Published, JBC Papers in Press, June 6, 2006, DOI 10.1074/jbc.M603085200 Akhilesh K. Singh 1 , Jayati Mullick, John Bernet, and Arvind Sahu 2 From the National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India Herpesvirus saimiri (HVS) is a lymphotropic virus that causes T-cell lymphomas in New World primates. It encodes a struc- tural homolog of complement control proteins named comple- ment control protein homolog (CCPH). Previously, CCPH has been shown to inhibit C3d deposition on target cells exposed to complement. Here we have studied the mechanism by which it inactivates complement. We have expressed the soluble form of CCPH in Escherichia coli, purified to homogeneity and com- pared its activity to vaccinia virus complement control protein (VCP) and human complement regulators factor H and soluble complement receptor 1. The expressed soluble form of CCPH bound to C3b (K D 19.2 M) as well as to C4b (K D 0.8 M) and accelerated the decay of the classical/lectin as well as alternative pathway C3-convertases. In addition, it also served as factor I cofactor and supported factor I-mediated inactivation of both C3b and C4b. Time course analysis indicated that although its rate of inactivation of C4b is comparable with VCP, it is 14-fold more potent than VCP in inactivating C3b. Site-directed mutagenesis revealed that Arg-118, which corresponds to Lys- 120 of variola virus complement regulator SPICE (a residue crit- ical for its enhanced C3b cofactor activity), contributes signifi- cantly in enhancing this activity. Thus, our data indicate that HVS encodes a potent complement inhibitor that allows HVS to evade the host complement attack. The complement system is an integral participant in the innate mechanisms of immunity and, thus, has a burden of per- forming surveillance in the host and protecting it from all the pathogens including viruses (1, 2). Earlier studies have deci- sively demonstrated that both acute and latent viruses are sus- ceptible to complement-mediated neutralization (3, 4). Thus, complement exerts a strong selective pressure on viruses dur- ing infection. These data suggest that for their successful sur- vival, viruses must have developed mechanisms to subvert this system. Consistent with this premise, genome sequencing of poxviruses and herpesviruses have shown that members of these families encode for structural homologs of human regu- lators of the complement activation (RCA) 3 family (5–9). The RCA family members are formed by tandemly repeating complement control protein (CCP) domains or short consen- sus repeats, which fold into a bead-like structure, and multiple CCPs are separated by linkers of 2–7 residues (10 –12). These proteins regulate complement by two distinct mechanisms (i) by accelerating the irreversible dissociation of the classical/lec- tin (C4b,2a) and alternative (C3b,Bb) pathway C3-convertases and (ii) by serving as cofactors in serine protease factor I-medi- ated inactivation of C3b and C4b (the subunits of C3-converta- ses) (13, 14). To date detailed characterization of all these activ- ities has been performed for the complement control protein homologs of vaccinia virus (VCP) (15–17), variola virus (SPICE) (18), monkeypox virus (MOPICE) (9), and Kaposi’s sarcoma- associated herpesvirus (Kaposica/KCP) (19, 20). Sequence comparison of the viral homologs of RCA (vCCPs) show that the sequence similarity between the pox- virus homologs exceeds 91%, whereas that among the her- pesvirus homologs varies from 43 to 89%. These data suggest that the herpesvirus homologs are more diverse in structure compared with the poxvirus homologs. Whether this struc- tural diversity in herpesvirus homologs is also reflected in their function is not clear, as among the herpesvirus homologs, detailed functional characterization has been per- formed only for the Kaposi’s sarcoma-associated herpesvirus homolog (Kaposica/KCP) (19 –22). Herpesvirus saimiri (HVS), the prototype of rhadinoviruses, is regularly found in its natural host, the squirrel monkey. Although it does not cause any disease in its natural host, infec- tion in other New World primates such as tamarins, common marmosets, and owl monkey causes acute peripheral T cell lym- phoma within less than 2 months (23, 24). In addition, the virus is also capable of transforming simian and human T cells in vitro (25, 26). Interestingly, unlike any other viruses, the HVS harbors two homologs of complement regulatory proteins, (i) a * This work was supported by the Wellcome Trust Senior Research Fellowship in Biomedical Science in India (to A. S.). The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. S The on-line version of this article (available at http://www.jbc.org) contains supplemental Figs. 1 and 2. 1 Supported by a research fellowship from the Council of Scientific and Indus- trial Research, India. This work was performed in partial fulfillment of a Ph.D. thesis, which will be submitted to the University of Pune, Pune, India. 2 To whom correspondence should be addressed. Tel.: 91-20-2569-0922; Fax: 91-20-2569-2259; E-mail: [email protected]. 3 The abbreviations used are: RCA, regulators of complement activation; AP, alternative pathway; CP, classical pathway; HVS, herpesvirus saimiri; CCP, complement control protein; vCCP, viral CCP; CCPH, complement control protein homolog; sCCPH, soluble CCPH of herpesvirus saimiri; VCP, vaccinia virus complement control protein; SPICE, smallpox inhibitor of complement enzymes; MOPICE, monkeypox inhibitor of complement enzymes; Kaposica, Kaposis’s sarcoma-associated herpesvirus inhibitor of complement activation; sCR1, soluble complement receptor 1; EA, anti- body coated sheep erythrocytes; ORF, open reading frame; RU, response unit. THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 32, pp. 23119 –23128, August 11, 2006 © 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A. AUGUST 11, 2006 • VOLUME 281 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 23119 by guest on December 17, 2020 http://www.jbc.org/ Downloaded from

Upload: others

Post on 26-Aug-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

Functional Characterization of the ComplementControl Protein Homolog of Herpesvirus SaimiriARG-118 IS CRITICAL FOR FACTOR I COFACTOR ACTIVITIES*□S

Received for publication, March 31, 2006, and in revised form, June 7, 2006 Published, JBC Papers in Press, June 6, 2006, DOI 10.1074/jbc.M603085200

Akhilesh K. Singh1, Jayati Mullick, John Bernet, and Arvind Sahu2

From the National Centre for Cell Science, Pune University Campus, Ganeshkhind, Pune 411007, India

Herpesvirus saimiri (HVS) is a lymphotropic virus that causesT-cell lymphomas in New World primates. It encodes a struc-tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH hasbeen shown to inhibit C3d deposition on target cells exposed tocomplement. Here we have studied the mechanism by which itinactivates complement.We have expressed the soluble form ofCCPH in Escherichia coli, purified to homogeneity and com-pared its activity to vaccinia virus complement control protein(VCP) and human complement regulators factor H and solublecomplement receptor 1. The expressed soluble form of CCPHbound toC3b (KD �19.2�M) aswell as toC4b (KD �0.8�M) andaccelerated the decay of the classical/lectin as well as alternativepathway C3-convertases. In addition, it also served as factor Icofactor and supported factor I-mediated inactivation of bothC3b and C4b. Time course analysis indicated that although itsrate of inactivation of C4b is comparable with VCP, it is 14-foldmore potent than VCP in inactivating C3b. Site-directedmutagenesis revealed that Arg-118, which corresponds to Lys-120 of variola virus complement regulator SPICE (a residue crit-ical for its enhanced C3b cofactor activity), contributes signifi-cantly in enhancing this activity. Thus, our data indicate thatHVS encodes a potent complement inhibitor that allowsHVS toevade the host complement attack.

The complement system is an integral participant in theinnatemechanisms of immunity and, thus, has a burden of per-forming surveillance in the host and protecting it from all thepathogens including viruses (1, 2). Earlier studies have deci-sively demonstrated that both acute and latent viruses are sus-ceptible to complement-mediated neutralization (3, 4). Thus,complement exerts a strong selective pressure on viruses dur-ing infection. These data suggest that for their successful sur-vival, viruses must have developed mechanisms to subvert thissystem. Consistent with this premise, genome sequencing of

poxviruses and herpesviruses have shown that members ofthese families encode for structural homologs of human regu-lators of the complement activation (RCA)3 family (5–9).The RCA family members are formed by tandemly repeating

complement control protein (CCP) domains or short consen-sus repeats, which fold into a bead-like structure, and multipleCCPs are separated by linkers of 2–7 residues (10–12). Theseproteins regulate complement by two distinct mechanisms (i)by accelerating the irreversible dissociation of the classical/lec-tin (C4b,2a) and alternative (C3b,Bb) pathway C3-convertasesand (ii) by serving as cofactors in serine protease factor I-medi-ated inactivation of C3b and C4b (the subunits of C3-converta-ses) (13, 14). To date detailed characterization of all these activ-ities has been performed for the complement control proteinhomologs of vaccinia virus (VCP) (15–17), variola virus (SPICE)(18), monkeypox virus (MOPICE) (9), and Kaposi’s sarcoma-associated herpesvirus (Kaposica/KCP) (19, 20).Sequence comparison of the viral homologs of RCA

(vCCPs) show that the sequence similarity between the pox-virus homologs exceeds 91%, whereas that among the her-pesvirus homologs varies from 43 to 89%. These data suggestthat the herpesvirus homologs are more diverse in structurecompared with the poxvirus homologs. Whether this struc-tural diversity in herpesvirus homologs is also reflected intheir function is not clear, as among the herpesvirushomologs, detailed functional characterization has been per-formed only for the Kaposi’s sarcoma-associated herpesvirushomolog (Kaposica/KCP) (19–22).Herpesvirus saimiri (HVS), the prototype of rhadinoviruses,

is regularly found in its natural host, the squirrel monkey.Although it does not cause any disease in its natural host, infec-tion in other New World primates such as tamarins, commonmarmosets, and owlmonkey causes acute peripheral T cell lym-phomawithin less than 2months (23, 24). In addition, the virusis also capable of transforming simian and human T cellsin vitro (25, 26). Interestingly, unlike any other viruses, theHVSharbors two homologs of complement regulatory proteins, (i) a

* This work was supported by the Wellcome Trust Senior Research Fellowshipin Biomedical Science in India (to A. S.). The costs of publication of thisarticle were defrayed in part by the payment of page charges. This articlemust therefore be hereby marked “advertisement” in accordance with 18U.S.C. Section 1734 solely to indicate this fact.

□S The on-line version of this article (available at http://www.jbc.org) containssupplemental Figs. 1 and 2.

1 Supported by a research fellowship from the Council of Scientific and Indus-trial Research, India. This work was performed in partial fulfillment of aPh.D. thesis, which will be submitted to the University of Pune, Pune, India.

2 To whom correspondence should be addressed. Tel.: 91-20-2569-0922; Fax:91-20-2569-2259; E-mail: [email protected].

3 The abbreviations used are: RCA, regulators of complement activation;AP, alternative pathway; CP, classical pathway; HVS, herpesvirus saimiri;CCP, complement control protein; vCCP, viral CCP; CCPH, complementcontrol protein homolog; sCCPH, soluble CCPH of herpesvirus saimiri;VCP, vaccinia virus complement control protein; SPICE, smallpox inhibitorof complement enzymes; MOPICE, monkeypox inhibitor of complementenzymes; Kaposica, Kaposis’s sarcoma-associated herpesvirus inhibitor ofcomplement activation; sCR1, soluble complement receptor 1; EA, anti-body coated sheep erythrocytes; ORF, open reading frame; RU, responseunit.

THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 281, NO. 32, pp. 23119 –23128, August 11, 2006© 2006 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in the U.S.A.

AUGUST 11, 2006 • VOLUME 281 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 23119

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 2: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

homolog of RCA encoded by ORF4 and (ii) a homolog of theterminal complement inhibitor CD59 encoded by ORF15 (27,28). The ORF4 was predicted to encode a protein containingfour CCP modules followed by a transmembrane domain.Analysis of posttranscriptional processing indicated that ORF4transcript occurs as unspliced as well as single-spliced mRNA.The unspliced mRNA codes for a membrane-bound glycopro-tein containing four extracellular CCPs alongwith a transmem-brane region, whereas the spliced mRNA codes for a solubleprotein that lacks transmembrane region (29).Initial characterization of the RCA homolog of HVS (named

complement control protein homolog, CCPH) showed thatexpression of themembrane form of this protein on BALB/3T3cells inhibited C3d deposition on these cells when they wereincubated with whole human serum (30). Although this studydemonstrated the complement inhibiting activity of this pro-tein, the mechanism by which it inactivates complement acti-vation was not elucidated. In the present study we describe themechanism of complement regulation by the RCA homolog ofHVS. Our results show that the soluble form of the RCA hom-olog (sCCPH; CCP1–4 without the transmembrane domain)interacts with complement proteins C3b as well as C4b andaccelerates decay of the classical/lectin and alternative pathwayC3-convertases. In addition, the protein also has the ability toserve as factor I cofactor and support factor I-mediated inacti-vation of C3b and C4b. Importantly, we show that its factor Icofactor activity for C3b is 14-fold higher in comparison toVCP, the most completely characterized vCCP, and that Arg-118 plays a critical role in enhancing this activity.

EXPERIMENTAL PROCEDURES

Reagents and Buffers—Antibody-coated sheep erythrocytes(EA) were made by incubating sheep erythrocytes with anti-sheep erythrocyte antibodies procured from ICN BiomedicalInc. (Irvine, CA). Veronal-buffered saline (VBS) contained 5mM barbital, 145 mM NaCl, and 0.02% sodium azide, pH 7.4.GVB was VBS containing 0.1% gelatin, GVB2� was GVB con-taining 0.5 mMMgCl2 and 0.15 mMCaCl2, and GVBE was GVBwith 10 mM EDTA.MgEGTA contained 0.1 MMgCl2 and 0.1 MEGTA, and phosphate-buffered saline, pH 7.4, contained 10mM sodium phosphate and 145 mM NaCl.Complement Proteins and Their Proteolytically Activated

Products—The human complement protein C3 was purifiedaccording to Hammer et al. (31) with minor modifications aspreviously described (17), and native C3was separated fromC3(H2O) by running on a Mono S column (32). The complementfactors H and I were kindly provided by Prof. Michael K. Pang-burn (University of Texas Health Centre, Tyler, TX.). Humanfactor B was purified as follows. Human plasma was subjectedto a stepwise precipitationwith 11 and 26% polyethylene glycol.The 26% polyethylene glycol precipitate was dissolved in 10mMsodium phosphate, pH 7.4, run on Source Q column (Amer-sham Biosciences) in the same buffer, and eluted with a lineargradient of 0–0.5 M NaCl. Fractions containing factor B wereidentified by Ouchterlony analysis, pooled, and loaded onto aMono S 5/5 column (Amersham Biosciences) in 50mM sodiumphosphate, pH6.0. Boundproteinswere elutedwith a linear saltgradient of 0–0.5 M NaCl and analyzed by SDS-PAGE. Homo-

geneous factor B fractions were pooled and concentrated. Therecombinant human soluble formof complement receptor type1 (sCR1) was a generous gift from Dr. Henry Marsh (AVANTImmunotherapeutics, Inc., Needham, MA.). C3b, the proteo-lytically activated form of C3, was generated by limited trypticcleavage of C3 and purified on a Mono Q 5/5 (Amersham Bio-sciences) column as previously described (16). C4b, the proteo-lytically activated form of C4, was purchased fromCalbiochem.Purity of all the proteins exceeded 95%, as judged by SDS-PAGEanalysis.Cloning, Expression, Purification, andRefolding of the Soluble

Form of Herpesvirus Saimiri Complement Control ProteinHomolog (sCCPH) and the R118A Mutant—The herpesvirussaimiri CCPH gene (CCP domains 1–4) was PCR-amplifiedfrom the CCPH clone pCEX-1 (a kind gift of Drs. John Lambris,Department of Pathology and LaboratoryMedicine, Universityof Pennsylvania, Philadelphia, PA and Jens-Christian Albrecht,Institut fur Klinische undMolekulare Virologie, Erlangen, Ger-many) with specific primers 5�-GGAATTCAGCTGTCCTA-CACGTAACCAG-3� (the EcoRI site is underlined) and5�-CCGCTCGAGCATACATTCAGGAATAGCTGG-3� (theXhoI site is underlined) and cloned into the bacterial expres-sion vector pET29 at the EcoRI and XhoI sites. The R118Amutant was constructed from this clone by using theQuikChange II site-directed mutagenesis kit (Stratagene, LaJolla, CA). After verifying the fidelity of both the clones byDNAsequencing, they were transformed into Escherichia coli BL21cells for expression.Expression of sCCPH and R118A mutant (numbering

according to the mature protein sequence (29)) was performedas described below. A single colony of the bacterial cloneexpressing sCCPH or the mutant protein was inoculated into 5ml of LB-kanamycin media (LB media containing 30 �g/mlkanamycin) and grown overnight at 37 °C, and 2 ml of this cul-ture was transferred into 100 ml of LB-kanamycin. The culturewas grown for 2 h at 37 °C, and thereafter 10 ml of the culturewas transferred to 600 ml of LB-kanamycin and grown at 37 °Cuntil the optical density reached 0.6 atA600. Protein expressionwas induced by the addition of 1 mM isopropyl 1-thio-�-D-ga-lactopyranoside, and the induced culture was further grown for4 h. The cells were harvested by centrifugation at 8000 rpm at4 °C.For purification of the expressed proteins, frozen cell pellets

(�16 g)were gently resuspended in 48ml of 50mMTris, pH8.0,1 mM EDTA, 100 mM NaCl, and 0.1 mM phenylmethylsulfonylfluoride. The cell suspension was then treated with lysozyme(0.3 mg/ml), stirred for 20 min, mixed with deoxycholic acid(1.3 mg/g), and stored at 37 °C for 30min. After incubation, thelysate was sonicated with 15 pulses of 15 s each and centrifugedat 10,000 � g for 20 min at 4 °C. The pellet containing theinclusion bodies was washed twice with 50 mM Tris, pH 8.0, 10mM EDTA, 100 mM NaCl, and 0.5% Triton X-100 and solubi-lized in 50mMTris, pH 8.0, 1mM EDTA, 100mMNaCl, and 8 Murea. The sample was then centrifuged at 4 °C for 30 min at11,000 rpm, and the supernatant obtained was loaded onto anickel nitrilotriacetic acid-agarose column (Qiagen, Hilden,Germany) pre-equilibrated with 100 mM NaH2PO4, 10 mMTris, 8 M urea, pH 8.0. The columnwaswashedwith the binding

Functional Characterization of HVS CCPH

23120 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 32 • AUGUST 11, 2006

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 3: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

buffer containing 10 mM imidazole, and the bound protein waseluted with 400 mM imidazole.

The purified proteins were refolded by using a rapid dilutionmethod (1:50) described previously (33). In brief, the purifiedprotein was added dropwise with continuous stirring into arefolding buffer containing 0.02 M ethanolamine, 1 mM EDTA,0.5 M L-arginine, 1 mM reduced glutathione, and 1 mM oxidizedglutathione, pH 11.0. The sample was then left static for 36 h.The refolded sample was concentrated, dialyzed against phos-phate-buffered saline, and subjected to SDS-PAGE, circulardichroism, and sequencing and mass analysis by mass spec-trometry (17).Measurement of Factor I Cofactor Activity—Analysis of

factor I cofactor activities of sCCPH and themutant was essen-tially performed as described (34). These assays were per-formed in physiological ionic strength buffer (phosphate-buff-ered saline).Measurement of Decay-accelerating Activity—The classical

pathway (CP) decay-accelerating activity of sCCPH and theR118A mutant were determined by forming EAC142 (35), andthe alternative pathway (AP) C3-convertase decay-acceleratingactivity was measured by forming C3b,Bb on sheep (ES) as wellas rabbit (ER) erythrocytes. The details of these methods havebeen described previously (17, 36).Circular Dichroism (CD)—The sCCPH and its mutant

R118Awere subjected toCD spectra in the farUV region (190–360 nm) using a Jasco J18 spectropolarimeter with a cylindricalquartz cell with a path length of 0.01 cm. The resolution was 1nm, the sensitivity was 20 millidegrees, and the speed was 10nm/min. Each presented spectrum is the measure of eightmeasurements. The concentration of both the proteins was 200�g/ml in 10mMphosphate containing 145mMNaCl, pH7.4.Allthe data were subtracted against the background data using thespectral analysis software.Flow Cytometry for Measurement of Inhibition of C3b

Deposition—Inhibition of the classical and alternative pathway-mediated C3b deposition on erythrocytes by sCCPH and VCPwas measured by flow cytometry (19). For measurement of theclassical pathway-mediated C3b deposition, 5 �l of EA (109/mlin GVB2�) was mixed with 2 �l of C8-deficient human serum(Calbiochem) and 2 �M sCCPH or VCP in a total volume of 44�l and incubated for 30 min at 37 °C. The cells were washedwith GVB2�, centrifuged, mixed with 100 �l of 1⁄100-diluted flu-orescein isothiocyanate-conjugated F(ab�)2 anti-C3 goat IgG(Cappel Laboratories, Warrington, PA), and further incubatedon ice for 1 h.After incubation, the cellswerewashed twicewith400 �l of GVB, resuspended in 1.0 ml of the same buffer, andanalyzed on a FACS Vantage (BD Biosciences). For measure-ment of alternative pathway-mediated deposition of C3b, 5 �lof rabbit erythrocytes (109/ml in GVB) was mixed with 2 �l of0.1 M MgEGTA, 3 �l of C8-deficient human serum (Calbio-chem), and 30 �l of GVB or GVB containing 2 �M sCCPH orVCP and incubated for 30 min at 37 °C. The cells were washedwith GVB, and deposition of C3b was detected as describedabove. Results are expressed as mean channel fluorescence of10,000 cells.Surface Plasmon Resonance Measurements—The kinetics of

sCCPH and the R118A mutant binding to C3b and C4b was

determined on the surface plasmon resonance-based biosensorBIACORE 2000 (Biacore AB, Uppsala, Sweden). The experi-ments were performed in phosphate-buffered saline-Tween(10 mM sodium phosphate, 145 mM NaCl, pH 7.4, containing0.05% Tween 20) at 25 °C. For proper orientation of these pro-teins, the free SH groups of bothC3b andC4bwere biotinylatedand then immobilized on the streptavidin chip (Sensor ChipSA, Biacore AB) (34). FC-2 was immobilized with C3b (1592RU), FC-3 was immobilized with C4b (1197 RUs), and FC-1(blank flow cell) served as the control flow cell. Because sCCPHshowed very little binding to C3b, more C3b molecules weredeposited onto FC-2 by forming C3-convertase (37, 38). Inbrief, �6000 RUs of C3b were deposited using three cycles ofC3b deposition. In each cycle, theC3-convertasewas formed byinjecting a mixture of factors B and D (5 �g of B and 0.35 �g ofD) and then 45 �g of native C3 was injected using the co-injectoption. Deposition of C3b onto the chipwas performed in vero-nal-buffered saline containing 1mMNiCl2. Formeasurement ofbinding of sCCPH and the mutant protein to C3b and C4b,various concentrations of these proteins were injected for 120 sat 50 �l/min. Dissociation was measured for 180 s. The sensorchips were regenerated with 30-s pulses of 0.2 M sodium car-bonate, pH 9.5. Sensograms obtained for the control flow cell(FC-1) were subtracted from the data for the flow cell immobi-lized with C3b or C4b, and the surface plasmon resonance dataobtained were evaluated by BIAevaluation software version 4.1using global fitting.

RESULTS

Expression and Characterization of sCCPH—Because a largequantity of protein was required for conducting multiple

FIGURE 1. Characterization of purified expressed herpesvirus saimiri-sCCPH and R118A mutant. Top left, SDS-PAGE analysis. Purified sCCPHand R118A mutants were separated on a 10% SDS-PAGE gel under reduc-ing conditions and stained with Coomassie Blue. Lane 1, molecular mass(MW) markers; lane 2, inclusion bodies containing sCCPH; lane 3, purifiedsCCPH; lane 4, inclusion bodies containing R118A mutant; lane 5, purifiedR118A mutant. Top right, CD spectra for sCCPH and R118A mutant. Bottom,mass analysis of sCCPH and R118A mutant. The molecular weights weredetermined by running the proteins on SDS-PAGE and by matrix-assistedlaser desorption ionization-time of flight mass spectrometry (17).

Functional Characterization of HVS CCPH

AUGUST 11, 2006 • VOLUME 281 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 23121

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 4: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

assays, we chose to expressHVS sCCPH in E. coli using the pETexpression system. The soluble form of HVS CCPHwas ampli-fied from the HVS clone pCEX-1 and cloned into the expres-sion vector pET29. The expressed protein was purified tohomogeneity using histidine affinity (Fig. 1), and the identity ofthe expressed protein was confirmed by sequencing usingmassspectrometry. The amino acid sequence of the expressed pro-tein was consistent with the predicted sequence confirming theidentity; the sequence coverage obtained was 86%. Theexpressed protein was refolded according to the methoddescribed by R. A. Smith and co-workers (33). This methodtypically provided a 20% yield. Analysis of the refolded proteinby circular dichroism yielded a peak around 230 nm, which is acharacteristic of CCP domains (39) (Fig. 1). These data con-firmed proper folding of the protein. The expressed proteinwas�95% pure as judged by SDS-PAGE analysis, and it migrated asa single band of 32,000 Da on the gel. Further mass analysis bymass spectrometry confirmed that its molecular mass was sim-ilar to its calculated mass (within error �1%) (Fig. 1).Previously it has been shown that the membrane form of

CCPH inhibits C3d deposition on the target cells (30). To verifyif the refolded protein is biologically active, we tested its abilityto inhibit C3b deposition on erythrocytes during complementactivation. As depicted in Fig. 2, sCCPH inhibited both the clas-sical as well as alternative pathway-mediated deposition of C3bon erythrocytes. Importantly, the data indicated that sCCPHwas more active than VCP in inhibiting the alternative path-way-mediated deposition of C3b.Kinetic Analysis of Interaction of sCCPH with Complement

Proteins C3b and C4b—The humancomplement control proteins inac-tivate complement by targeting C3band/or C4b. Because sCCPH inhib-ited C3b deposition mediated byboth the classical and alternativepathways, we sought to analyze itsinteraction with C3b and C4b bysurface plasmon resonance technol-ogy. In this assay, C3b and C4bwereimmobilized in their physiologicalorientation on a streptavidin chip bylabeling their free SH groups withbiotin (34), and sCCPHwas injectedover the chip to measure binding.The sCCPH showed good bindingtoC4b but veryweak binding toC3b(Fig. 3, upper left panel). Bindingdata obtained by injecting variousconcentrations of sCCPH fittedwellto 1:1 bindingmodel (ka � 158; kd �5.32 � 10�3; KD � 3.35 � 10�5;�2 � 0.273). Because the bindingresponse was very low, we furtherdeposited C3b on the sensor chip toincrease the response and reevalu-ate affinity. More C3b was depos-ited by forming AP C3-convertaseon the chip and flowing native C3

FIGURE 2. Inhibition of C3b deposition on erythrocytes during comple-ment activation by sCCPH and VCP. Top panel, classical pathway-medi-ated C3b deposition on erythrocytes was measured by incubating EA andC8-deficient human serum with or without 2 �M sCCPH/VCP at 37 °C for 30min. Deposition of C3b was detected by fluorescence-activated cell sort-ing using fluorescein isothiocyanate-conjugated F(ab�)2 anti-C3 goatimmunoglobulin G. Bottom panel, alternative pathway-mediated C3bdeposition on erythrocytes was measured by incubating rabbit erythro-cytes and C8-deficient human serum in the presence of MgEGTA with orwithout 2 �M sCCPH/VCP at 37 °C for 20 min. Deposition of C3b wasdetected as described above. Control samples contained 10 mM EDTA.

FIGURE 3. Surface plasmon resonance analysis of binding of sCCPH to complement proteins C3b and C4b.Biotinylated C3b and C4b were oriented in their physiological orientation on a streptavidin chip (Sensor Chip SA;Biacore AB), and various concentrations of sCCPH were injected over the chip to measure binding. Top left, bindingof sCCPH to C3b and C4b oriented on a streptavidin chip by labeling their free SH groups with biotin (34)). Top right,deposition of C3b by forming AP C3-convertase on the chip (37, 38). Factors B and D mix and C3 (in veronal-bufferedsaline containing 1.0 mM NiCl2) were repeatedly injected over the chip using the co-inject option of Biacore 2000.Bottom left, sensogram overlay for the interaction between sCCPH and C3b deposited using the AP C3-convertase.The solid lines represent the global fitting of the data to a 1:1 Langmuir binding model (A � B7AB; BIAevaluation4.1). The concentration of sCCPH injected is indicated at the right of the sensograms. Bottom right, sensogramoverlay for the interaction between sCCPH and C4b. The arrow indicates the time point used for evaluating thesteady-state affinity data. The concentration of sCCPH injected is indicated at the right of the sensograms.

Functional Characterization of HVS CCPH

23122 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 32 • AUGUST 11, 2006

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 5: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

(37, 38). Three cycles of AP amplification resulted in depositionof�6000 RUs of C3b (Fig. 3, upper right panel); non-covalentlyassociatedBb andC3bwere removed by injection of brief pulsesof 0.2 M sodium carbonate, pH 9.5. As expected, deposition ofC3b using this approach resulted in a decaying surface, suggest-ing that C3b was attached to the surface by forming ester link-ages (40). When sCCPH was flown over the enzyme-coupledC3b, it showed good binding response (Fig. 3, lower left panel).To calculate affinities for C3b and C4b, various concentra-

tions of sCCPHwere injected over the chip. The sCCPH boundto both C3b and C4b in a dose-dependent manner (Fig. 3).Global fitting analysis of the sensograms showed a good fit ofsCCPH-C3b data to the 1:1 binding model with a drifting baseline (�2 � 1.83), but sCCPH-C4b data could not be fitted to 1:1model, and therefore, it was evaluated by steady-state analysis.These data indicated that sCCPH-C3b interaction follows asimple 1:1 binding model, whereas sCCPH-C4b interaction iscomplex. A comparison of the affinities showed that sCCPHhas a 24-fold higher affinity for C4b than C3b (Table 1).sCCPH Acts as a Cofactor in Factor I-mediated Cleavage of

C3b andC4b—Thedata presented above indicated that sCCPHinteracted with C3b as well as C4b (Fig. 3). We, therefore, ana-lyzed the ability of sCCPH to serve as a cofactor in factor I-me-diated cleavage of C3b andC4b. A fluid phase assay was utilized

for determining the factor I cofactor activity wherein C3b orC4b was incubated with factor I and sCCPH or the controlproteins (VCP, factor H, or sCR1), and cleavages of the�� chainwere assessed by running the samples on SDS-PAGE gels. Dur-ing factor I-mediated cleavage of C3b, the �� chain of C3b wascleaved at three distinct sites depending on the cofactorinvolved. In the assay utilized, the appearance of 68- and46-kDa fragments indicated cleavage at site 1 and generation ofiC3b1, the appearance of 68- and 43-kDa fragments indicatedcleavages at sites 1 and 2 and generation of iC3b2, and theappearance of 43-kDa, C3dg, and 25-kDa fragments indicatedthe cleavage at all the three sites and generation of C3c andC3dg. In the case of C4b, factor I is known to cleave at two sites.The appearance ofC4d and 25-and 16-kDa fragments indicatedboth these cleavages and generation of C4c and C4d.It is clear from the data presented in Fig. 4 that sCCPH acted

as a factor I cofactor in mediating cleavages of C3b as well asC4b. The data further showed that its cofactor activity for C3bdiffered from VCP in two ways; (i) it was much more efficientthan VCP in supporting C3b cleavages, and (ii) unlike VCP,which primarily supported factor I cofactor activity for the firstsite, it displayed efficient cofactor activity for both sites 1 and 2,leading to generation of iC3b2. In comparison to human com-plement regulators, its cleavage pattern was similar to factor H,but it differed from sCR1 in that it did not support the thirdcleavage. The cofactor activity of sCCPH for C4b was similar toVCP and sCR1, and like these proteins it also supported thecleavages at both the sites and led to generation of C4c andC4d.Because sCCPH showed greater factor I cofactor activity for

C3b than VCP (Fig. 4), we studied C3b inactivation as a func-tion of time using equimolar concentrations of different cofac-tors (Fig. 5). The data indicated that the time required for 50%cleavage of the�� chain of C3b for sCCPHwas 2.5min, whereasthat for VCP was 34 min, indicating that sCCPH was about a14-fold more efficient cofactor than VCP. The sCCPH, how-ever, was less efficient compared with human complement reg-ulators factorH and sCR1; the time required for 50% cleavage ofthe �� chain of C3b in the presence of factor H and sCR1 was

1.25 and 1.0 min, respectively. Asimilar time course experiment per-formed for C4b cleavage showedthat sCCPH was �2-fold less effi-cient than VCP (Fig. 6). The timerequired for 50% cleavage of the ��chain of C4b for sCCPH, VCP, andsCR1 was 27, 14.5, and 3.5 min,respectively.sCCPHAccelerates theDecay of the

Classical and Alternative PathwayC3-convertases—Thedecay-accelera-tionactivityof sCCPHfor theclassicaland alternative pathway C3-conver-tases was measured by hemolyticassays. For measuring the CP decay-accelerating activity, sensitized sheeperythrocytes coated with the C3-con-vertase enzyme (C4b,2a) was incu-batedwith sCCPHor the control pro-

FIGURE 4. Factor I cofactor activity of sCCPH, VCP, factor H (fH), and sCR1 for complement proteins C3band C4b. Cofactor activity for C3b cleavage was assayed by mixing 3 �g of C3b, 0.1 �g of factor I, and 1.5 �g ofthe cofactor (as indicated in the lanes) in 20 �l of physiologic ionic strength buffer and incubating at 37 °C forthe indicated time periods. Cofactor activity for C4b cleavage was performed by mixing 3 �g of C4b, 0.1 �g offactor I, and 1 �g of the cofactor (as indicated in the lanes) in 20 �l and incubating at 37 °C for the indicated timeperiods. Cleavage products were visualized by separating the samples on SDS-PAGE gel (9.5% for C3b and 10%for C4b) under reducing conditions and staining with Coomassie Blue.

TABLE 1Kinetic and affinity data for the interactions of sCCPH, R118Amutant, and VCP with human complement proteins C3b and C4bka, association rate constant; kd, dissociation rate constant; KD, equilibrium rateconstant; NA, not applicable.

Ligand Analyte kd/ka S.E. (kd/ka) KD �2

s�1/M�1 M

C3b sCCPH 2.63 � 10�3/137 2.05 � 10�5/3.39 1.92 � 10�5 1.83aC4b sCCPH NA NA 8.04 � 10�7 2.87bC3b R118A 4.06 � 10�3/18.7 4.75 � 10�5/0.73 2.17 � 10�4 9.44aC4b R118A NA NA 9.64 � 10�7 1.23bC3b VCP NA NA 2.0 � 10�6 1.47bC4b VCP NA NA 3.65 � 10�7 1.12b

a Data were calculated by global fitting to a 1:1 Langmuir binding model(BIA evaluation 4.1).

b Data were calculated by steady-state analysis (BIA evaluation 4.1).

Functional Characterization of HVS CCPH

AUGUST 11, 2006 • VOLUME 281 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 23123

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 6: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

teins, and the enzyme decay was quantitated by measuringhemolysis after adding EDTA sera. sCCPH showed good decay-accelerating activity and decayed 83% enzyme activity at 1�M, buton a molar basis it was 5.8-fold less active than VCP and 18-foldless active than sCR1 (Fig. 7A).The APC3-convertase decay-accelerating activity was quan-

titated by forming the enzyme (C3b,Bb) on sheep or rabbiterythrocytes and incubating it with sCCPH or the control pro-tein. In comparison to VCP, sCCPH showed 8-fold more decayactivity for the enzyme present on sheep cells (Fig. 7B). It was,however, much less efficient compared with human comple-ment regulators factor H and sCR1. Previously it has beenshown that VCP does not decay the AP C3-convertase presenton rabbit cells (17). To see if this also holds true for sCCPH, wetested its decay activity for the enzyme present on rabbit eryth-rocytes. Our results showed that unlike VCP it also decays the

enzyme present on rabbit erythrocytes. The concentrationrequired for 50% decay was 12.5 �M, which was 1.6-fold morecompared with the enzyme present on sheep cells.Arginine 118 of sCCPH Is Crucial for Its Cofactor Activities

but Not Decay-accelerating Activities—Because sCCPHshowed a robust factor I cofactor activity for C3b cleavage, wesought to identify the determinant that is responsible for itsenhanced activity. Recent studies on SPICE has shown that itpossesses �100-fold more factor I cofactor activity for C3bthan VCP (18), and residues Lys-108 and -120 are primarilyresponsible for this enhanced cofactor activity (41). Sequencealignment of sCCPH with SPICE showed that sCCPH has Leu(Leu-106) and Arg (Arg-118) in the corresponding positions.Because earlier studies have shown that ionic interactions playa critical role in CCP-C3b/C4b interactions (34, 42–44) we sus-pected thatArg-118might be responsible for the enhancedC3bcofactor activity. To further probe this possibility, we built athree-dimensional model of sCCPH by homology modelingusing the crystal structure of VCP (45) as the template. Themodel structure showed thatArg-118 is exposed to solvent (Fig.8), which further supported the possibility that this residuecould be involved in the cofactor activity.To study the involvement of Arg-118 in the functional activ-

ities of sCCPH, we mutated the Arg-118 to Ala and expressedthe R118A mutant in E. coli using the pET expression system.Purification and refolding procedures used for themutant wereessentially similar to that of sCCPH. Sequencing (sequence cov-erage obtained was 91%) and circular dichroism analysis con-firmed the identity and correct folding of the mutant (Fig. 1),respectively. Functional analysis of the R118A mutant showedthat its factor I cofactor activity for C3b was drasticallydecreased compared with sCCPH; it was 50-fold less activecompared with sCCPH (Fig. 9). Interestingly, the mutant alsoshowed a 12-fold decrease in factor I cofactor activity for C4b(Fig. 9).Next, we analyzed if this mutation also affects the decay-

accelerating activities of sCCPH. The data showed that themutant was as active as sCCPH in accelerating the decay of theclassical as well as alternative pathwayC3-convertases (Fig. 10).Together these data indicated that Arg-118 plays an importantrole in enhancing the cofactor activities but not the decay-ac-celerating activities of sCCPH.Binding of R118A Mutant to C3b and C4b—Binding of the

complement control proteins to C3b and C4b is a prerequi-site for imparting factor I cofactor activities and decay-ac-celerating activities; however, a significant body of literaturesuggests that binding does not always correlate well withthese activities (14, 46). To determine whether R118A muta-tion affected binding, we measured binding of this mutant toC3b and C4b using the surface plasmon resonance assay. TheR118A mutant showed about an 11-fold decrease in affinityfor C3b compared with sCCPH (Table 1), which was a resultof a 7.3-fold decrease in the on-rate and a 1.5-fold increase inthe off-rate. These data are consistent with the substantialdecrease in C3b cofactor activity. There was, however, nodecrease in the affinity of C4b (Table 1), although themutantshowed a 12-fold decrease in C4b cofactor activity. Further-more, it is clear from the data provided in Fig. 10 that R118A

FIGURE 5. Time course of factor I cofactor activity of sCCPH, VCP, factor H(fH), and sCR1 for complement protein C3b. Cofactor activity was meas-ured by incubating 3 �g of C3b with 0.05 �g of factor I and 0.5 �M concentra-tions of the cofactors (as indicated in each gel) at 37 °C for the indicated timeperiod. The reactions were stopped by adding the sample buffer containingdithiothreitol, and the cleavage products were visualized by separating thesamples on 9.5% SDS-PAGE gel and staining with Coomassie Blue. The inten-sities of the �� chain were determined by densitometric analysis and repre-sented graphically (lower panel).

Functional Characterization of HVS CCPH

23124 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 32 • AUGUST 11, 2006

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 7: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

mutation had no effect on the decay-accelerating activities;hence, the decrease in KD value for C3b did not correlatewith the AP C3-convertase activity.

DISCUSSION

The regulators of human complement belonging to the RCAfamily contain 4–59 copies of CCP domains. It is believed thatthe sequence variations imposed on the CCP domain fold andthe interdomain dynamics determine the differences in func-tionality of the complement regulators (8, 11, 47–49). Inviruses, homologs of complement regulators have beendescribed in members of Herpesviridae and Poxviridae. Thesequence similarity among the poxviral complement regulatorsexceeds 91%, whereas that among the herpesviral complementregulators ranges between 43 and 89%. Therefore, functionalcharacterization of various herpesviral complement regulatorswould help in determining whether structural diversity in theseregulators has led to any change in their functional diversity.Functional complement regulators in Herpesviridae familyhave been described in herpesvirus saimiri (30), �-herpesvirus68 (50), and Kaposi’s sarcoma-associated herpesvirus (KSHV)(19, 20), but detailed functional analysis for decay-acceleratingactivities, factor I cofactor activities, and binding to C3b andC4b have been performed only for the KSHV complement reg-ulator (Kaposica/KCP) (19–21). In the present study we haveanalyzed the functional activities of HVS sCCPH to get insightinto the functional diversity of sCCPH against the complementsystem.A previous study had shown that themembrane formofHVS

CCPH inhibits the classical pathway-mediated deposition ofC3d onto the target cells (30). Our data on inhibition of C3bdeposition onto the target cells by sCCPH show that it inhibitsboth the classical as well as alternative pathway-mediated dep-osition of C3b (Fig. 2). These results are consistent with theprevious data on herpesviral (�-HV68 and Kaposica) and pox-viral (VCP and SPICE) complement regulators (16,18–20, 50),which showed inhibitory activities against both the pathways.Earlier, using hemolytic assays it has been shown that vCCPsare efficient in inactivating the classical pathway (IC50 � 0.1–0.2�M) (16, 19); these values are considered significant becausethe local concentration of these proteins at the site of infectionis expected to be very high (2, 20, 51). Measurement of inhibi-tion of theCP-mediated lysis of sheep erythrocytes showed thatlike other vCCPs, sCCPH is also an effective inhibitor of theclassical pathway (IC50 � 0.27 �M). Further analysis of the CPC3-convertase regulatory activities demonstrated that it con-tains both effective factor I cofactor activity for C4b (Fig. 6) aswell as CP decay-accelerating activity (Fig. 7A). Thus, like othervCCPs, the effective classical pathway inhibitory activity is alsoconserved in HVS CCPH.It is clear from the data presented in Fig. 2 that sCCPH is

significantly more active than VCP in inhibiting the alternativepathway-mediated deposition of C3b onto erythrocytes. Todefine themechanism responsible for this increased activity, wecharacterized its factor I cofactor activity for C3b and decay-accelerating activity for the AP C3-convertase. The datarevealed that sCCPH possesses 14-fold more C3b cofactoractivity compared with VCP (Fig. 5). In fact, the cofactor activ-ity was only about 2.5-fold less compared with human comple-ment regulators factor H and sCR1 (Fig. 5). We would like topoint out here that the observed difference in the cofactor activ-

FIGURE 6. Time course of factor I cofactor activity of sCCPH, VCP, and sCR1for complement protein C4b. Cofactor activity was measured by incubating3 �g of C4b with 0.1 �g of factor I and 1 �M concentrations of the cofactor (asindicated in each gel) at 37 °C for the indicated time periods. The reactionswere stopped by adding the sample buffer containing dithiothreitol, and thecleavage products were visualized by separating the samples on 10% SDS-PAGE gel and staining with Coomassie Blue. The intensities of the �� chainwere determined by densitometric analysis and are represented graphically(lower panel).

Functional Characterization of HVS CCPH

AUGUST 11, 2006 • VOLUME 281 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 23125

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 8: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

ity of sCCPH compared with VCPwas not due to the differencein affinity for C3b as sCCPH showed lower affinity for C3bcompared with VCP (Table 1). This, however, is not surprising

as previously it has been shown thatCD46, which has a much loweraffinity for C3b than CR1, has ahigher cofactor activity than CR1(52). It is likely that the increasedcofactor activity of sCCPH is aresult of its better interaction withfactor I. Analysis of AP C3-conver-tase decay-accelerating activityshowed that sCCPH is a poor decayaccelerator of AP C3-convertase(Fig. 7B). Although its activity was8-fold better compared with VCP, itwas �2000-fold less active com-pared with factor H and sCR1 (Fig.7B). Together these data suggestthat the increased alternative path-way inhibitory activity of sCCPHwas primarily due to its increasedfactor I cofactor activity for C3b.

Factor I is known to cleave C3b at three different positionsdepending on the cofactors involved: the first between 1281–1282, which generates iC3b1; the second between 1298–1299,which generates iC3b2; the third between 932–933, which gen-erates C3c and C3dg. Whether vCCPs support the cleavage ofC3b to C3c and C3d/C3dg was under debate until recently (18,20), but it is now clear that viral regulators primarily supportthe cleavage of C3b to iC3b1 (e.g. VCP (9, 16)) or iC3b2 (e.g.Kaposica (19, 22), SPICE (9, 41), andMOPICE (9)) and not C3cand C3dg. Like most of other viral regulators, the sCCPH alsosupported the generation of C3b to iC3b2 (Figs. 4 and 5).Because generation of iC3b1 itself is sufficient to inactivate C3b(16), it is not clear whether inactivation of C3b to iC3b2 asopposed to iC3b1 provides any functional advantage to viruses.

Because sCCPH showed about a 14-fold higher cofactoractivity for C3b, we sought to examine the basis for thisincreased activity. Earlier, Rosengard et al. (18) demonstratedthat SPICE is about 100-fold more potent than VCP in inacti-vating C3b. Later, using the site-directed mutagenesisapproach, it was established that Lys-108 and -120 residues areprincipally responsible for better functioning of SPICE (41).When we aligned sCCPH sequence with SPICE to determinewhether sCCPHcontains positively charged residues at the cor-responding positions, we found that sCCPH contains Leu (Leu-106) and Arg (Arg-118) at these positions. Based on these, wepredicted that Arg-118 might be responsible for the highercofactor activity of sCCPH. Modeling of the sCCPH structurebased on the crystal structure of VCP demonstrated that theside chain of Arg-118 is exposed to solvent (Fig. 8), which fur-ther supported this possibility. Thus, we mutated the Arg-118to Ala and examined its functional activities. We found thatremoval of charge at this position drastically affected the factorI cofactor activity for C3b (50-fold decrease) and to some extentC4b (12-fold decrease) but had no effect on the decay-acceler-ating activities (Figs. 9 and 10). These data along with the pre-vious studies on SPICE clearly point out that the presence of apositive charge at this position enhances theC3b cofactor activ-ity in viral homologs. It is interesting to note that despite

FIGURE 7. Decay acceleration of the classical and alternative pathway C3-convertase by sCCPH, VCP, factorH, and sCR1. A, the CP C3-convertase (C4b,2a) was formed on sheep erythrocytes and incubated with variousamounts of sCCPH or the control proteins (VCP, factor H, and sCR1) for 5 min at 22 °C in DGVB2�. The remainingC3-convertase activity was measured by hemolysis after the addition of 1:100 diluted guinea pig serum containing20 mM EDTA. B, the AP C3-convertase (C3b,Bb) was formed on sheep erythrocytes and incubated with sCCPH or thecontrol proteins (VCP, factor H, and sCR1) for 10 min at 37 °C. The remaining C3-convertase activity was measured byhemolysis after the addition of 1:10 diluted human sera containing 20 mM EDTA. The data were normalized bysetting 100% C3-convertase activity to be equal to the average activity in the absence of inhibitor.

FIGURE 8. Model of sCCPH. The model was built by utilizing the crystal struc-ture of VCP (Ig40) (45) as the template using SWISS-MODEL (54 –56). A, over-lap of ribbon models of sCCPH (red) and VCP (green) structures. The sidechains of Arg-118 of sCCPH and Glu-120 of VCP are labeled. B, solid surfacepresentation of sCCPH model showing exposed Arg-118 in blue.

Functional Characterization of HVS CCPH

23126 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 32 • AUGUST 11, 2006

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 9: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

belonging to two different viral families, substitution of posi-tively charged residue at comparable positions has been seen inSPICE as well as sCCPH (Lys-120 in SPICE and Arg-118 insCCPH). Although a previous study on SPICE (41) examinedthe role of Lys-120 in enhancingC3b cofactor activity, it did notlook at its role either in C4b cofactor activity or in decay-accel-erating activities. Based on our data, we suggest that Lys-120 ofSPICE might also play a role in enhancing its cofactor activityfor C4b.In summary, our data clearly show thatHVSCCPHpossesses

all the complement regulatory activities present in Kaposicaand other viral regulators. Thus, it seems that despite signifi-

cant sequence differences between herpesviral complementregulators, the functional activities have been conserved. Thesedata along with previous observations, therefore, point out thatmaintenance of various complement regulatory functionsmustbe important to the pox as well as herpesviruses and inhibitionof the lectin/classical pathway is crucial to viral survival thanthe inhibition of alternative pathway. Whether sequence varia-tions in herpesviral complement proteins have resulted inacquisition of any new functions is not clear at present andrequires further studies. Previously, it has been demonstratedthat CCP homolog of �HV-68 plays an important role in com-plement evasion in vivo (53). Given the fact that sCCPH is anefficient complement inactivator, it is likely that sCCPH mayalso act as an immune evasion molecule in vivo and protectHVS from the host complement during infection.

Acknowledgments—We thank Drs. John D. Lambris (Department ofPathology and Laboratory Medicine, University of Pennsylvania,Philadelphia, PA) and Jens-Christian Albrecht (Institut fur KlinischeundMolekulare Virologie, Erlangen, Germany) for providing theHVSCCPH clone, Dr. Michael K. Pangburn (Department of Biochemistry,University of Texas Health Science Center, Tyler, Texas) for supportand the generous gift of complement proteins factors D, H, and I, Dr.HenryMarsh (AVANT Immunotherapeutics, Inc., Needham,Ma) forproviding sCR1, Dr. Nicholas E. Sherman (Biomolecular ResearchFacility, University of Virginia, Charlottesville) for protein sequencingand mass analysis, and Dr. M. V. Krishnasastry (National Centre forCell Science, Pune) for generating the sCCPHmodel, Dr. K. N. Ganesh(National Chemical Laboratory, Pune) for access to his spectropola-rimeter. We also express appreciation to Yogesh Panse, Sha-ranabasava Hallihosur, and Hemangini Shikhare for excellent tech-nical assistance.

REFERENCES1. Cooper, N. R. (1998) in The Human Complement System in Health and

Disease (Volanakis, J. E., and Frank, M. M., eds) pp. 393–407, MarcelDekker, Inc., New York

2. Mullick, J., Kadam, A., and Sahu, A. (2003) Trends Immunol. 24, 500–5073. Bernet, J., Mullick, J., Singh, A. K., and Sahu, A. (2003) J. Biosci. 28,

249–2644. Lachmann, P. J. (2002) Proc. Natl. Acad. Sci. U. S. A. 99, 8461–84625. Seet, B. T., Johnston, J. B., Brunetti, C. R., Barrett, J. W., Everett, H., Cam-

eron, C., Sypula, J., Nazarian, S. H., Lucas, A., and McFadden, G. (2003)Annu. Rev. Immunol. 21, 377–423

6. Alcami, A., and Koszinowski, U. H. (2000) Trends Microbiol. 8, 410–4187. Means, R. E., Lang, S.M., Chung, Y. H., and Jung, J. U. (2002) Front. Biosci.

7, 185–2038. Gewurz, B. E., Gaudet, R., Tortorella, D., Wang, E. W., and Ploegh, H. L.

(2001) Curr. Opin. Immunol. 13, 442–4509. Liszewski, M. K., Leung, M. K., Hauhart, R., Buller, R. M., Bertram, P.,

Wang, X., Rosengard, A. M., Kotwal, G. J., and Atkinson, J. P. (2006)J. Immunol. 176, 3725–3734

10. Hourcade, D., Holers, V. M., and Atkinson, J. P. (1989)Adv. Immunol. 45,381–416

11. Kirkitadze, M. D., and Barlow, P. N. (2001) Immunol. Rev. 180, 146–16112. Blom, A.M., Villoutreix, B. O., andDahlback, B. (2004)Mol. Immunol. 40,

1333–134613. Pangburn, M. K. (2000) Immunopharmacology 49, 149–15714. Lambris, J. D., Sahu, A., andWetsel, R. (1998) inTheHumanComplement

System in Health and Disease (Volanakis, J. E., and Frank, M., eds) pp.83–118, Marcel Dekker Inc., New York

15. Mckenzie, R., Kotwal, G. J., Moss, B., Hammer, C. H., and Frank, M. M.

FIGURE 9. Time course of factor I cofactor activity of R118A mutant forcomplement proteins C3b and C4b. Cofactor activity was measured byincubating C3b or C4b with factor I and R118A mutant at 37 °C for the indi-cated time period. The reactions were stopped by adding the sample buffercontaining dithiothreitol, and the cleavage products were visualized by sep-arating the samples on SDS-PAGE gels and staining with Coomassie Blue. Theintensities of the �� chain were determined by densitometric analysis andrepresented graphically (lower panel).

FIGURE 10. Comparison of the classical and alternative pathway C3-con-vertase decay-accelerating activity of sCCPH and R118A mutant. Sheeperythrocytes coated with the classical or alternative pathway C3-convertaseswere incubated with sCCPH or the R118A mutant, and the remaining enzymeactivity was measured by incubating the cells with EDTA-sera as describedunder “Experimental Procedures.”

Functional Characterization of HVS CCPH

AUGUST 11, 2006 • VOLUME 281 • NUMBER 32 JOURNAL OF BIOLOGICAL CHEMISTRY 23127

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 10: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

(1992) J. Infect. Dis. 166, 1245–125016. Sahu, A., Isaacs, S. N., Soulika, A.M., and Lambris, J. D. (1998) J. Immunol.

160, 5596–560417. Mullick, J., Bernet, J., Panse, Y., Hallihosur, S., Singh, A. K., and Sahu, A.

(2005) J. Virol. 79, 12382–1239318. Rosengard, A. M., Liu, Y., Nie, Z., and Jimenez, R. (2002) Proc. Natl. Acad.

Sci. U. S. A. 99, 8808–881319. Mullick, J., Bernet, J., Singh, A. K., Lambris, J. D., and Sahu, A. (2003)

J. Virol. 77, 3878–388120. Spiller, O. B., Blackbourn, D. J., Mark, L., Proctor, D. G., and Blom, A. M.

(2003) J. Biol. Chem. 278, 9283–928921. Mullick, J., Singh, A. K., Panse, Y., Yadav, V., Bernet, J., and Sahu, A. (2005)

J. Virol. 79, 5850–585622. Mark, L., Lee, W. H., Spiller, O. B., Proctor, D., Blackbourn, D. J.,

Villoutreix, B.O., and Blom,A.M. (2004) J. Biol. Chem. 279, 45093–4510123. Desrosiers, R. C., and Falk, L. A. (1982) J. Virol. 43, 352–35624. Desrosiers, R. C., Bakker, A., Kamine, J., Falk, L. A., Hunt, R. D., and King,

N. W. (1985) Science 228, 184–18725. Biesinger, B., Trimble, J. J., Desrosiers, R. C., and Fleckenstein, B. (1990)

Virology 176, 505–51426. Cabanillas, J. A., Cambronero, R., Pacheco-Castro, A., Garcia-Rodriguez,

M. C.,Martin-Fernandez, J.M., Fontan, G., and Regueiro, J. R. (2002)Clin.Exp. Immunol. 127, 366–373

27. Albrecht, J. C., Nicholas, J., Biller, D., Cameron, K. R., Biesinger, B., New-man, C., Wittmann, S., Craxton, M. A., Coleman, H., and Fleckenstein, B.(1992) J. Virol. 66, 5047–5058

28. Means, R. E., Choi, J. K., Nakamura, H., Chung, Y. H., Ishido, S., and Jung,J. U. (2002) Curr. Top Microbiol. Immunol. 269, 187–201

29. Albrecht, J. C., and Fleckenstein, B. (1992) J. Virol. 66, 3937–394030. Fodor,W. L., Rollins, S. A., Biancocaron, S., Rother, R. P., Guilmette, E. R.,

Burton, W. V., Albrecht, J. C., Fleckenstein, B., and Squinto, S. P. (1995)J. Virol. 69, 3889–3892

31. Hammer, C. H., Wirtz, G. H., Renfer, L., Gresham, H. D., and Tack, B. F.(1981) J. Biol. Chem. 256, 3995–4006

32. Pangburn, M. K. (1987) J. Immunol. Methods 102, 7–1433. White, J., Lukacik, P., Esser, D., Steward, M., Giddings, N., Bright, J. R.,

Fritchley, S. J., Morgan, B. P., Lea, S. M., Smith, G. P., and Smith, R. A.(2004) Protein Sci. 13, 2406–2415

34. Bernet, J.,Mullick, J., Panse, Y., Parab, P. B., and Sahu, A. (2004) J. Virol. 78,

9446–945735. Pan, Q., Ebanks, R. O., and Isenman, D. E. (2000) J. Immunol. 165,

2518–252736. Pryzdial, E. L., and Isenman, D. E. (1986)Mol. Immunol. 23, 87–9637. Jokiranta, T. S., Hellwage, J., Koistinen, V., Zipfel, P. F., andMeri, S. (2000)

J. Biol. Chem. 275, 27657–2766238. Harris, C. L., Abbott, R. J., Smith, R. A.,Morgan, B. P., and Lea, S.M. (2005)

J. Biol. Chem. 280, 2569–257839. Kirkitadze, M. D., Krych, M., Uhrin, D., Dryden, D. T., Smith, B. O., Coo-

per, A., Wang, X., Hauhart, R., Atkinson, J. P., and Barlow, P. N. (1999)Biochemistry 38, 7019–7031

40. Sahu, A., and Pangburn, M. K. (1994) J. Biol. Chem. 269, 28997–2900241. Sfyroera, G., Katragadda, M., Morikis, D., Isaacs, S. N., and Lambris, J. D.

(2005) J. Immunol. 174, 2143–215142. Blom, A. M., Webb, J., Villoutreix, B. O., and Dahlback, B. (1999) J. Biol.

Chem. 274, 19237–1924543. Krych, M., Hauhart, R., and Atkinson, J. P. (1998) J. Biol. Chem. 273,

8623–862944. Oran, A. E., and Isenman, D. E. (1999) J. Biol. Chem. 274, 5120–513045. Murthy, K.H., Smith, S. A., Ganesh,V. K., Judge, K.W.,Mullin,N., Barlow,

P. N., Ogata, C. M., and Kotwal, G. J. (2001) Cell 104, 301–31146. Pangburn, M. K., Pangburn, K. L., Koistinen, V., Meri, S., and Sharma,

A. K. (2000) J. Immunol. 164, 4742–475147. Stehle, T., and Larvie, M. (2003) in Innate Immunity (Ezekowitz, R. A. B.,

and Hoffmann, J. A., eds) pp. 221–253, Humana Press Inc., Totowa, NJ48. Zhang, L., and Morikis, D. (2006) Biophys. J. 90, 3106–311949. Krych-Goldberg, M., and Atkinson, J. P. (2001) Immunol. Rev. 180,

112–12250. Kapadia, S. B.,Molina, H., van, B. V., Speck, S. H., and Virgin, H.W. (1999)

J. Virol. 73, 7658–767051. Kotwal, G. J. (2000) Immunol. Today 21, 242–24852. Seya, T., Okada, M., Nishino, H., and Atkinson, J. P. (1990) J. Biochem.

(Tokyo) 107, 310–31553. Kapadia, S. B., Levine, B., Speck, S. H., and Virgin, H.W. (2002) Immunity.

17, 143–15554. Schwede, T., Kopp, J., Guex, N., and Peitsch, M. C. (2003) Nucleic Acids

Res. 31, 3381–338555. Guex, N., and Peitsch, M. C. (1997) Electrophoresis 18, 2714–272356. Peitsch, M. C. (1995) Bio/Technology 13, 658–660

Functional Characterization of HVS CCPH

23128 JOURNAL OF BIOLOGICAL CHEMISTRY VOLUME 281 • NUMBER 32 • AUGUST 11, 2006

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from

Page 11: FunctionalCharacterizationoftheComplement ... · tural homolog of complement control proteins named comple-ment control protein homolog (CCPH). Previously, CCPH has ... evade the

Akhilesh K. Singh, Jayati Mullick, John Bernet and Arvind SahuACTIVITIES

Herpesvirus Saimiri: ARG-118 IS CRITICAL FOR FACTOR I COFACTOR Functional Characterization of the Complement Control Protein Homolog of

doi: 10.1074/jbc.M603085200 originally published online June 7, 20062006, 281:23119-23128.J. Biol. Chem. 

  10.1074/jbc.M603085200Access 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

Supplemental material:

  http://www.jbc.org/content/suppl/2006/06/08/M603085200.DC1

  http://www.jbc.org/content/281/32/23119.full.html#ref-list-1

This article cites 52 references, 26 of which can be accessed free at

by guest on Decem

ber 17, 2020http://w

ww

.jbc.org/D

ownloaded from