SlideShare a Scribd company logo
1 of 9
Download to read offline
JOURNAL OF VIROLOGY, Jan. 2004, p. 683–691 Vol. 78, No. 2
0022-538X/04/$08.00ϩ0 DOI: 10.1128/JVI.78.2.683–691.2004
Copyright © 2004, American Society for Microbiology. All Rights Reserved.
The Spacing between Cysteines Two and Three of the LDL-A Module
of Tva Is Important for Subgroup A Avian Sarcoma and
Leukosis Virus Entry
Tia Rai, Deborah Marble, Kayla Rihani, and Lijun Rong*
Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612
Received 17 July 2003/Accepted 3 October 2003
Rong et al. have demonstrated previously that with a few substitutions, the fourth repeat of human
low-density lipoprotein (hLDL-A4) receptor can functionally replace the LDL-A module of Tva, the cellular
receptor for subgroup A avian sarcoma and leukosis virus (ASLV-A), in viral entry (L. Rong, K. Gendron, and
P. Bates, Proc. Natl. Acad. Sci. USA 95:8467-8472, 1998). Here we have shown that swapping the amino
terminus of hLDL repeat 5 (hLDL-A5) with that of Tva, in addition to the corresponding substitutions made
in human LDL-A4, was required to convert hLDL-A5 into an efficient ASLV-A receptor. These results sub-
stantiated our previous findings regarding the role of the specific residues in the viral interaction domain of
Tva and demonstrated the critical role of the amino terminus of the Tva LDL-A module in ASLV-A infection.
Furthermore, we have shown that the residues between cysteines 2 and 3 of the Tva LDL-A module in a
Tva/LDL-A5 chimeric protein can be functionally replaced by the corresponding region of another LDL-A
module, human LDL receptor-related protein repeat 22 (LDL-A22), to mediate efficient ASLV-A entry. Since
the only conserved feature between the C2-C3 region of LDL-A22 and the Tva LDL-A module is that both
contain nine amino acids of which none are conserved, we conclude that the spacing between C2 and C3 of the
LDL-A module of Tva is an important determinant for ASLV-A entry. Thus, the present study provides strong
evidence to support our hypothesis that one role of the N terminus of the LDL-A module of Tva is to allow
proper folding and conformation of the protein for optimal interaction with the viral glycoprotein EnvA in
ASLV-A entry.
Entry of subgroup A avian sarcoma and leukosis virus
(ASLV-A) into its hosts is mediated by interactions between
the viral glycoprotein EnvA and a small cell surface glycopro-
tein, Tva (1, 29). Direct interaction of EnvA with Tva is not
only important for initial viral attachment but is also important
for the postbinding steps in ASLV-A entry. Binding between
Tva and EnvA in vitro can induce a series of conformational
changes on EnvA that lead to exposure of the putative fusion
peptide and its association with the lipid membrane (7, 11, 13,
19). These results provide direct biochemical evidence that
receptor binding can trigger the structural rearrangements
likely required for membrane fusion mediated by a viral gly-
coprotein. Since receptor triggering appears to be a common
yet poorly understood mechanism for entry of various envel-
oped viruses, analysis of the Tva-EnvA interaction serves as an
excellent model in elucidating the basic principles in viral en-
try.
One of the major advantages of using Tva/EnvA as a model
to elucidate the viral entry mechanism is that the viral inter-
action domain of Tva is solely determined by a single low-
density lipoprotein (LDL) receptor module within the extra-
cellular domain of Tva (3, 21), making it a simple model
amenable to molecular, biochemical, and structural analysis.
The LDL-A module of Tva is 40 amino acids in length and
includes six invariable cysteines and five highly conserved
acidic residues found in other LDL-A modules such as human
LDL receptor and LDL receptor-related proteins (1). Bio-
chemical and structural analysis of different LDL-A modules
demonstrates that the six conserved cysteines are involved in
the formation of three pairs of disulfide bonds. Furthermore,
structural analysis of several individual LDL-A modules (and,
recently, of the entire ectodomain of human LDL receptor) by
X-ray crystallography showed that the side chains of four con-
served acidic residues near the C terminus and the carbonyl
oxygen groups of two nonacidic residues of each LDL-A mod-
ule coordinate calcium binding (9, 24, 25). These common
structural features of LDL-A modules are important for
proper folding and, thus, for their functions in ligand binding.
The role of the Tva LDL-A module in EnvA binding and
ASLV-A entry has been extensively examined by molecular,
biochemical, and structural analysis. It has been demonstrated
that the LDL-A module of Tva efficiently mediated ASLV-A
entry when it was appended to a heterologous membrane-
spanning domain (21). Furthermore, this module of Tva can be
functionally replaced by a modified human LDL-A4 module in
mediation of ASLV-A entry (22). These studies, together with
mutational analysis (23, 31, 32), have identified several puta-
tive viral interaction residues important for viral entry. Bio-
chemical and structural analysis demonstrated that like other
LDL-A modules, the correct in vitro folding of the Tva LDL-A
module is calcium dependent (26–28). Surprisingly, however,
calcium is not essential for EnvA binding once the protein is
correctly folded (30).
* Corresponding author. Mailing address: Department of Microbi-
ology and Immunology, College of Medicine, University of Illinois at
Chicago, E829 MSB, 835 S. Wolcott Ave., Chicago, IL 60612. Phone:
(312) 355-0203. Fax: (312) 996-6415. E-mail: lijun@uic.edu.
683
One important structural difference between the Tva and
other LDL-A modules is that the Tva LDL-A module does
not have the signature anti-parallel ␤-sheet observed at the
N-terminal C1-C3 region of other LDL-A modules (5, 6, 8,
9, 17, 20, 25, 26, 28). The C2-C3 region of the Tva LDL-A
module contains nine residues instead of the four to five
residues seen in the other LDL-A modules, and this region
of Tva is responsible for the flexible conformation at the N
terminus (28). In this study, we wanted to examine the role
of this region in determining the receptor function of Tva.
We found that the spacing between the C2 and C3 regions
of the Tva LDL-A module is an important determinant for
ASLV-A entry. The present study provides strong evidence
that one role of the N-terminal region of the Tva LDL-A
module is to allow proper folding and overall conformation
of the protein for optimal interaction with EnvA in ASLV-A
entry.
MATERIALS AND METHODS
Cells and viruses. Human embryonic kidney 293T cells were maintained as
described previously (22). RCAS(A)AP (10) viral stocks were generated as
described previously (21). The EnvA-pseudotyped human immunodeficiency
virus (HIV) viral stocks were generated using the HIV vector pNL4-3.Luc.R-E
(4, 12) cotransfected with ASLV-A glycoprotein EnvA into 293T cells. Briefly,
293T cells were transiently transfected with 10 ␮g of the HIV vector either alone
or with 10 ␮g of EnvA DNA or vesicular stomatitis virus glycoprotein G
(VSV-G) DNA by a modified CaPO4 method as previously described (21). The
supernatants (viral stocks) were harvested 48 h posttransfection, filtered through
a 0.22-␮m-pore-size filter, and stored at Ϫ80°C.
Cloning, mutagenesis, and mutant nomenclature. A chimeric Tva construct
containing the human LDL-A5 instead of the Tva LDL-A module was generated
as follows. The PCR-amplified coding region for human LDL-A5 was digested
(using human LDL receptor cDNA as the template) (18) with restriction endo-
nucleases BamHI and SacII, and the digested DNA fragment was inserted into
the BamHI/SacII-digested Myc-Tva vector (22). This construct is referred to as
TL5, where T stands for Tva and L5 pertains to the LDL-A5 region. The other
TL5 derivatives were generated using the TL5 construct as the template by a
standard two-step PCR protocol. Four amino acids in the TL5 construct, namely,
FIG. 1. (A) Sequence alignment of quail Tva LDL-A module with human LDL-A4 and LDL-A5 receptors 4 and 5. Residues are numbered
according to the sequence of the mature Tva (1). The six invariable cysteines are boxed. The consensus amino acids among these and other LDL-A
modules are shown at the bottom. (B) Tva/hLDL-A5 constructs generated in this study. Human LDL-A5 (hLDL-A5) is 35 amino acids in length
and is numbered from 1 to 35. The four residues in LDL-A5 that were targeted are underlined. The nomenclature of the TL5 constructs is similar
to that used previously for the TL4 constructs.
684 RAI ET AL. J. VIROL.
R19, G23, E33, and G34, were replaced with the corresponding residues of the
Tva LDL-A module either individually or in combination (Fig. 1B). Two addi-
tional TL5 chimeric constructs were generated. TL5 chimera 1 has the N-
terminal C1-C3 region from the Tva LDL-A module and also contains the
substitutions R19L, G23H, and E33W. TL5 chimera 2 is same as TL5 chimera 1
except that it also has the N34G substitution (Fig. 1B). In addition, constructs C5
and L5 were generated using TL5 chimera 2 as the template but contained the
C2-C3 region from the chicken Tva LDL-A module and the C2-C3 region of the
human LDL-A22 module of LRP2, respectively.
Analysis of protein expression. Human kidney embryonic 293T cells were
transiently transfected with Myc-Tva and the TL5 constructs by a modified
CaPO4 method. Cells were lysed 48 h after transfection, and protein expression
was examined by Western blotting as previously described (22). Myc-tagged Tva
and TL5 proteins were detected using anti-myc monoclonal antibody (MAb)
9E10 as the primary antibody.
Protein surface expression was examined by fluorescence-activated cell sorter
(FACS) analysis. Briefly, 293T cells were transiently transfected with 10 ␮g of
DNAs of myc-Tva and TL5 constructs by the CaPO4 method. At 48 h posttrans-
fection, cells were resuspended in FACS buffer (phosphate-buffered saline con-
taining 2% fetal calf serum) to 5 ϫ 106
cells/ml. Cells were seeded into a
U-bottom 96-well plate and were incubated with 50 ␮l of mouse MAb 9E10 (10
␮g/ml stock) for 30 min. Cells were then washed twice with FACS buffer and
incubated with 50 ␮l of fluorescein isothiocyanate-conjugated rat anti-mouse
immunoglobulin G1 antibody at a final concentration of 10 ␮g/ml for 20 min.
Cells were washed again and resuspended in 300 ␮l of FACS buffer. Cells were
stained for viability with 0.5 ␮l of propidium iodide and were analyzed using a
Becton Dickinson FACSCalibur flow cytometer and CellQuest software.
ELISA-based binding test. To examine the ability of the TL5 chimeric proteins
to bind EnvA, an enzyme-linked immunosorbent assay (ELISA)-based binding
test, which was modified from a published protocol (22, 23), was developed.
Human 293T cells were transiently transfected with gD-EnvA, myc-Tva, and the
TL5 chimeric constructs shown in Fig. 1B. The transfected cells were induced
with 10 mM sodium butyrate 24 h posttransfection and lysed 40 h posttransfec-
tion with 1% Triton lysis buffer. Protein expression was examined and semiquan-
tified by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-
PAGE) and Western blotting using MAbs 9E10 (for myc-tagged Tva and TL5
constructs) and 1D3 (for gD-tagged EnvA). The ELISA binding test was per-
formed as follows: 96-well plates were coated with MAb 9E10 for 24 h. After
three washings, the plates were blocked for 30 min with phosphate-buffered
saline containing 0.05% Tween 20, 0.5% bovine serum albumin, and 0.05%
sodium azide. A total of 100 ml of lysates from cells transfected with wild-type
(wt) Tva or TL5 construct DNAs was added to each well for 1 h with shaking at
4°C. The plates were then washed three times with distilled water, and 100 ␮l of
gD-EnvA lysate was added to each well for 1 h with shaking. The wells were
washed and incubated with an anti-EnvA tail rabbit antibody for one more hour
at 4°C. Finally, the plates were washed four times, ABTS [2,2Ј-azinobis (3-
ethylbenzthiazolinesulfonic acid] and hydrogen peroxide were added to each
well, and the absorbance at 405 nm was read after incubation. Each experiment
was performed in triplicate, and the relative ability of each protein to bind EnvA
was expressed as the percentage of wt Tva binding to EnvA.
Infectivity assays. Two different viral systems were employed in this study: a
recombinant ASLV-A, RCAS(A)AP, which carries an alkaline phosphatase
(AP) gene as the reporter (10), and the EnvA-pseudotyped HIV viruses, which
carry a luciferase gene as the reporter (4, 12). The infectivity assay was carried
out as previously described (21). Briefly, 293T cells were transiently transfected
with DNAs of the Tva or TL5 constructs by the CaPO4 method. At 24 h
posttransfection, cells were seeded in six-well plates and then challenged with
RCAS(A)AP- or the EnvA-pseudotyped HIV viruses. The RCAS(A)AP-in-
fected cells were stained for AP, and the AP-positive cells were enumerated
under a microscope. The EnvA-pseudotyped HIV-infected cells were lysed, and
luciferase activity was measured with a standard luminometer.
RESULTS
Human LDL-A5 cannot replace the Tva LDL-A module for
viral receptor function. A chimeric Tva protein called TL5, in
which the Tva LDL-A module was replaced with human LDL
receptor repeat 5 (LDL-A5) (Fig. 1B), was created. To test its
ability to mediate viral entry, TL5 DNA was transiently trans-
fected into 293T cells and the transfected cells were challenged
with a recombinant ASLV-A carrying an AP reporter gene
[RCAS(A)AP)]. As expected, TL5 was unable to mediate viral
entry (data not shown). To ensure that the defect was not due
to a lack of protein expression, the TL5 transfected cells were
analyzed by Western blotting; it was found that total protein
expression was comparable to that of a myc-tagged wt Tva (Fig.
2, lanes 1 and 2). The surface expression was measured by flow
cytometry, and it was found indistinguishable from that of wt
Tva (data not shown). In addition, TL5 was also examined,
using an ELISA-based test (described later and in Materials
and Methods), for its ability to bind the ASLV-A glycoprotein
EnvA. As expected, TL5 was unable to bind EnvA (see Fig. 5).
These results are similar to the previous findings with TL4, a
chimeric protein between Tva and human LDL-A4 which was
shown to be unable to bind EnvA and which could not mediate
ASLV-A entry (22).
Human LDL-A5 with substitutions corresponding to the
viral interaction residues of Tva cannot be converted into an
effective viral receptor. Previously it was shown that a human
LDL-A4 module with a few amino acid substitutions could
functionally substitute for the Tva LDL-A module in media-
tion of ASLV-A entry (22). This gain-of-function approach was
instrumental in identifying several putative viral interaction
FIG. 2. Analysis of protein expression of TL5 constructs. Transient expression of the TL5 constructs was examined by Western blotting
following SDS-PAGE. At 48 h posttransfection, 293T cells were lysed with Triton lysis buffer and aliquots of samples were subjected to SDS-PAGE
and Western blotting. Molecular mass markers (in kilodaltons) are shown to the right of the panel.
VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 685
residues in Tva. Sequence comparison of human LDL-A5 with
human LDL-A4 and the Tva LDL-A module indicated that
none of the putative interaction residues of Tva, Leu34, His38,
Trp48, and Gly49 are conserved in LDL-A5 (Fig. 1A). To test
whether TL5 could be converted into a functional ASLV-A
receptor, 11 TL5 substitution variants were generated in which
the corresponding residues in TL5 were replaced with the four
putative viral interaction residues of Tva either individually or
in combination (Fig. 1B).
Human 293T cells were transiently transfected with the TL5
construct DNAs and lysed. Protein expression was examined
by Western blotting using 9E10, an antibody which specifically
recognizes the myc tag portion of these proteins. All 11 TL5
constructs were expressed well in 293T cells (Fig. 2, lanes 3, 4,
5, 7, 8, 9, and 11 to 15). As was previously observed with wt Tva
and TL4 constructs (22, 23), each of the TL5 proteins migrated
as a smear of multiple bands on SDS-PAGE, indicating that
these proteins were heavily modified by glycosylation in 293T
cells.
To examine the ability of the TL5 proteins to mediate
ASLV-A infection, the 293T cells transiently transfected with
the TL5 construct DNAs were challenged with the
RCAS(A)AP and the recombinant ASLV-A vector; the in-
fected cells were enumerated postinfection. Since expression
of Tva in native avian cells is extremely low, we were concerned
that overexpression of the TL5 constructs in 293T cells by
transient transfection might mask the defect of these proteins
in mediation of viral entry. To avoid this potential problem,
various amounts (1, 5, and 20 ␮g) of DNA of each TL5 con-
struct as well as wt Tva were used in transfection prior to
RCAS(A)AP challenges. Protein expression was detectable by
Western blotting and flow cytometry when 5 or 20 ␮g of DNA
was used in transfection (data not shown).
As shown in Fig. 3, challenging the wt Tva DNA-transfected
293T cells with RCAS(A)AP viruses resulted in high numbers
of AP-positive cells (approximately 105
AP-positive cells per
milliliter of viral stock used) when either a low or high amount
of DNA was used in transfection, indicating that wt Tva can
efficiently mediate ASLV-A entry. As a negative control,
mock-transfected 293T cells were also challenged with
RCAS(A)AP viruses; no background AP-positive cells were
detected. Among the 11 TL5 constructs, only three (TL5LHG,
TL5LHW, and TL5LHWG) mediated low levels of ASLV-A
infection at a level at least 100-fold lower than that seen with
wt Tva (Fig. 3). The rest of the TL5 constructs did not display
any viral receptor function (data not shown). These results are
in stark contrast with those seen with the TL4 chimeric con-
structs with respect to mediation of ASLV-A infection, as
previously reported (22). For example, while TL5LHWG was
approximately 100-fold lower than that seen with wt Tva, the
similar TL4 construct (TL4G-A19LD23H) was able to mediate
efficient viral entry. One plausible explanation is that human
LDL-A4 is more conserved than human LDL-A5 with respect
to the Tva LDL-A module between cysteines 3 and 6 (Fig. 1A).
These results suggest that additional viral interaction determi-
nants must be critical for maintaining the optimal viral recep-
tor function of Tva.
The amino terminus of the Tva LDL-A module is required to
convert human LDL-A5 into an efficient viral receptor. Previ-
ous reports suggest that the amino-terminal region of the Tva
LDL-A module plays a subtle role in mediation of ASLV-A
infection (22, 23). Thus, we generated two additional chimeric
constructs (named TL5 chimera 1 and TL5 chimera 2) which
replaced the human LDL-A5 C1-C3 region of TL5LHW and
TL5LHWG with the corresponding region of Tva (Fig. 1B).
These constructs were expressed well in 293T cells compared
to the other TL5 proteins (Fig. 2, lanes 6 and 10). TL5 chimera
1 mediated low levels of ASLV-A infection, while TL5 chimera
2 was as efficient as wt Tva in mediation of viral infection in
transfected 293T cells challenged with the RCAS(A)AP vi-
FIG. 3. The abilities of TL5 constructs to mediate ASLV-A entry as determined using RCAS(A)AP vector. Human 293T cells transiently
transfected with various amounts of plasmid DNA of the constructs shown in Fig. 1 were challenged with a series of 10-fold dilutions of
RCAS(A)AP, a recombinant ASLV-A virus carrying an AP reporter gene. Infected cells were fixed with 2% paraformaldehyde at 48 h
postinfection and stained for AP activity. The ability of a construct to mediate viral infection is expressed as the number of AP-positive cells (in
international units) per milliliter of virus stock used. Experiments were repeated several times with consistent results, and the results of a
representative experiment are shown here.
686 RAI ET AL. J. VIROL.
ruses (Fig. 3). These results provide strong evidence that the
region between cysteines 1 and 3 of the Tva LDL-A module
plays a crucial role in the viral receptor function of Tva. In
addition, since TL5 chimera 1 and TL5 chimera 2 differ by a
single residue (Gln in TL5 chimera 1 versus Gly in TL5 chi-
mera 2) (Fig. 1B), these results suggest that maintaining a
small residue-like glycine at this position is important for the
viral receptor function of the TL5 constructs, a finding fully
consistent with the previous report by Rong et al. of investi-
gations with human LDL-A4 (22).
Examination by an EnvA-pseudotyped HIV infection assay
of ASLV-A entry for TL5 constructs. The assay described
above, which uses a recombinant ASLV-A virus for infection
followed by AP staining and counting under a microscope for
infectivity, is a reliable method for measurement and compar-
ison of the abilities of Tva and its derivatives or mutants to
mediate viral entry. However, one major drawback of this assay
is that counting under a microscope is time consuming and
error prone. Thus, we used the luciferase gene as the reporter
to establish an HIV-based pseudotyping system to quantita-
tively measure ASLV-A entry.
To test the feasibility and specificity of this system, the DNA
of HIV vector pNL4-3.Luc.R-E (4, 12) was cotransfected with
the DNAs of either the ASLV-A glycoprotein EnvA or VSV-G
to generate the pseudotyped HIV viral stocks (see Materials
and Methods for the details). The produced viral stocks were
used to challenge mock-transfected human 293T cells or the
293T cells transiently expressing Tva, and the luciferase activity
of the cells was determined with a luminometer. The cells
infected by the VSV-G-pseudotyped HIV viral stock, as ex-
pected, gave a high level of luciferase activity (approximately 8
logs of relative light units [RLU]) regardless of whether Tva
was expressed or not (Fig. 4A), indicating that VSV-G medi-
ated entry is Tva independent. In contrast, the cells challenged
with the EnvA-pseudotyped HIV viruses only gave a high level
of luciferase activity (also about 8 logs of RLU) when the cells
expressed Tva. However, the cells challenged by the HIV viri-
ons lacking a glycoprotein, or the cells without Tva expression
challenged by the EnvA-pseudotyped HIV virions, only gave
background luciferase activity (Fig. 4A). These results indicate
that EnvA-mediated HIV entry is Tva dependent, as expected,
demonstrating the feasibility and specificity of the EnvA-
pseudotyped HIV system to measure ASLV-A entry.
The abilities of the TL5 chimeric constructs (shown in Fig. 3)
to mediate ASLV-A entry were also tested using the EnvA-
pseudotyped HIV viruses. TL5 chimera 2 could mediate
ASLV-A entry as efficiently as wt Tva, while TL5 chimera 1
was at least three logs less efficient than Tva. Furthermore,
TL5LHWG was better than TL5LHW, while TL5LHG was
unable to mediate ASLV-A entry (Fig. 4B). Thus, the results
were remarkably consistent with that seen with RCAS(A)AP
infection (Fig. 3). Therefore, we have established an easy and
quantitative assay to measure ASLV-A entry; this assay will
facilitate studies of Tva-EnvA interactions.
Most TL5 proteins do not efficiently bind to ASLV-A glyco-
protein EnvA. To further investigate the interaction between
the TL5 proteins and EnvA, the binding properties of these
proteins to EnvA were examined by an ELISA-based binding
test. This assay was modified from a previous protocol (23)
which allows quick assessment of a large number of Tva mu-
tants for their ability to bind EnvA. As shown in Fig. 5, among
the 14 TL5 proteins only TL5 chimera 2 displayed a relatively
high level of EnvA binding, giving approximately one-third of
that of wt Tva, while the remaining 13 TL5 proteins displayed
little or no detectable EnvA binding (less than 10% of that of
wt Tva). These results suggest that the failure of these TL5
constructs to mediate efficient ASLV-A infection is due to a
defect in high-affinity binding to EnvA. It is interesting that
although three TL5 constructs (TL5LHW, TL5LHWG, and
TL5 chimera 1) supported a low level of ASLV-A infection
(Fig. 3 and 4), they did not display detectable EnvA binding
(Fig. 5). Rong et al. have previously observed similar discrep-
ancies (measured by ELISA-based test) between the ability of
many Tva mutants and TL4 proteins to mediate viral entry and
FIG. 4. (A) Establishment (using a luciferase gene as the reporter) of the EnvA-pseudotyped HIV system. Human 293T cells were transiently
transfected with Tva or mock transfected, and the transfected cells were challenged with the EnvA-pseudotyped HIV viruses. The infected cells
were lysed, and luciferase activity levels were measured and are expressed as RLU. EnvA, the ASLV-A glycoprotein-pseudotyped HIV; vector,
HIV without any viral glycoprotein. Experiments (performed in triplicate) were repeated several times. The results of a representative experiment
are shown here. (B) The abilities of TL5 constructs to mediate ASLV-A entry as measured using the EnvA-pseudotyped HIV viruses. Mock, mock
transfected; TL5 Ch2, TL5 chimera 2; TL5 Ch1, TL5 chimera 1. Experiments (performed in triplicate) were repeated three times with consistent
results. Bars, standard deviations.
VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 687
their ability to bind EnvA (22, 23). A plausible explanation for
these discrepancies is that the ELISA-based binding test used
in this and previous studies is not sensitive enough to distin-
guish the binding differences among the TL5 proteins that have
low-level binding affinities to EnvA.
The spacing between the second and third cysteines of the
Tva LDL-A module is important for receptor function. Previ-
ously we have shown that the overall conformation of the Tva
LDL-A module is different from the structures of the other
reported LDL-A modules, including human LDL-A5. The N
terminus of the Tva LDL-A module does not contain the
signature antiparallel ␤-sheet observed in other LDL-A mod-
ules and is more flexible than other reported LDL-A modules
(28). Sequence alignment shows that there are nine residues
between the second and third cysteines for the Tva LDL-A
module and that the other LDL-A modules with reported
structures contain only four to six residues. The fact that the
TL5 chimera 2 can mediate ASLV-A infection efficiently and
TL5LHWG cannot do so prompted us to examine whether the
overall conformation of the N terminus of the Tva LDL-A
module contributes to the receptor function of Tva. As previ-
ously reported, LDL-A22 is the only other known LDL-A
module that (just like that of Tva) contains nine residues be-
tween C2 and C3, thus conserving the spacing between cys-
teines 2 and 3. Wang et al. proposed that LDL-A22 adopts a
structure similar to that of the Tva LDL-A module (28). How-
ever, none of the nine amino acids in the C2-C3 regions are
conserved between human LDL-A22 and the quail Tva LDL-A
module, while four of the nine residues are conserved between
quail and chicken Tva proteins.
Two TL5 variants, C5 and L5, which are identical to TL5
chimera 2 except that the residues between C2 and C3 of TL5
chimera 2 have been replaced with the corresponding residues
of chicken Tva (C5) or LDL-A22 of LDL receptor-related
protein (L5), respectively, were constructed (Fig. 6A). When
they were examined by FACS analysis, both constructs were
expressed well on the surface of the transfected 293T cells
(data not shown). The EnvA-pseudotyped HIV viruses were
used to challenge the transfected 293T cells as described above
to examine the TL5 variants for their ability to mediate
ASLV-A entry. The cells transiently expressing C5 gave a level
of luciferase activity comparable to that of wt Tva and TL5
chimera 2 (Fig. 6B), suggesting that C5 is as efficient as wt Tva
and TL5 chimera 2 in mediation of ASLV-A infection. Sur-
prisingly, the cells transiently expressing L5 gave a level of
luciferase approximately 100-fold higher than the cells express-
ing TL5LHWG and only about 6-fold lower than the Tva-
expressing cells (Fig. 6B). Since the amino acid sequences of
TL5LHWG and L5 are identical between C3 and C6 and are
very similar between C1 and C2, we can conclude that the
amino acids between C2 and C3 of L5 play an important role
in mediation of ASLV-A entry. Furthermore, since only the
spacing between C2 and C3 of human LDL-A22 is conserved
with respect to that of Tva from quail and chicken, we can
conclude that the proper spacing (and thus, likely the proper
conformation) of this region plays an important role for the
viral receptor function.
DISCUSSION
In this report we have shown that the putative viral interac-
tion residues of Tva that were previously identified by a gain-
of-function approach and mutational analysis are indeed crit-
ical for the conversion of human LDL-A5 into a functional
ASLV-A receptor. Furthermore, our results have revealed a
critical role of the N terminus between cysteines 2 and 3 of the
FIG. 5. The EnvA-binding abilities of TL5 constructs measured by an ELISA-based binding test. Human 293T cells were transiently transfected
with gD-EnvA, myc-Tva, and the TL5 chimeric constructs. An ELISA-based binding test (described in Materials and Methods) was performed to
measure the relative level of affinity of binding to EnvA of each of the TL5 proteins compared to that of myc-Tva. Mock, mock transfected.
688 RAI ET AL. J. VIROL.
Tva LDL-A module in mediation of ASLV-A entry. Signifi-
cantly, our results indicate that the spacing between C2 and C3
of Tva (and thus, the overall conformation of this region) is an
important determinant for viral entry.
The role of Tva in ASLV-A entry has been extensively ex-
amined by molecular, biochemical, and structural analysis.
However, most work has concentrated on the C-terminal re-
gion of the Tva LDL-A module (C3-C6); this region was found
to be essential for viral entry. This has led to the identification
of individual residues in this region which are either ligand
interaction residues or involved in stability and conformation
of the protein (2, 22, 23, 31, 32). For example, Leu34, His38,
Trp48, and Gly49 of Tva were identified as being important for
viral receptor function. These results are substantiated by the
results of the present study. We have shown that replacements
of the corresponding residues in human LDL-A5 by these four
amino acids, in addition to the amino terminus of Tva, were
required to convert human LDL-A5 into a functional viral
receptor. Interestingly, the side chains of His38 and Trp48 of
Tva were shown to be exposed on the surface, in consistency
with the notion that these residues are EnvA contact residues.
However, the side chain of Leu34 is buried in the interior of
the module together with Phe16 and Pro21, forming the hy-
drophobic core. These data indicate that the role of Leu34 is to
maintain the correct folding of Tva; thus, it is unlikely to be a
ligand contact residue (28). We speculate that conversion of
the corresponding residue of human LDL-A5 (Arg19) to a
leucine (as described in this study) or conversion of the corre-
sponding residue of human LDL-A4 (Ala19) to a leucine (as
described in the previous report) can help maintain the correct
folding in TL5 and TL4 chimeras, respectively.
The major difference between the present study using
human LDL-A5 and the previous study using LDL-A4 is
that LDL-A5 with the aforementioned four substitutions
was not able to mediate efficient viral infection unless its N
terminus was also replaced with the corresponding region
from Tva. This finding is in contrast to that of the previous
study with LDL-A4, which demonstrated that LDL-A4
could be converted into a functional viral receptor with
merely the aforementioned substitutions and without re-
placement of the N terminus by that of Tva (22). This may
be explained by a higher level of sequence homology be-
tween Tva and LDL-A4 than between Tva and LDL-A5 at
the C terminus. For example, there are two lysine residues
in LDL-A5 (K27 and K29) (Fig. 1A) that are absent in the
Tva module and LDL-A4. Perhaps these positively charged
residues adversely affect the binding affinity between
LDL-A5 and EnvA, thus making it a less efficient viral
receptor. It should be pointed out that the modified human
LDL-A4 protein (TL4G-A19LD23H) was also somewhat
defective in EnvA binding compared to that of wt Tva (22).
This defect in EnvA binding could be corrected by replacing
the N terminus of LDL-A4 with the corresponding region of
Tva (data not shown), again suggesting the important role of
the N-terminal region of Tva in mediation of efficient EnvA
binding and ASLV-A infection.
Sequence alignment between Tva and other LDL-A mod-
ules indicates that the N terminus of the Tva LDL-A module
(between cysteines 1 and 3) is more divergent than the C
terminus of other LDL-A modules (Fig. 1A). Indeed, the
structure of the LDL-A module of Tva reveals that compared
to the other LDL-A module structures reported to date, the N
terminus of the Tva module adopts a unique conformation.
The Tva LDL-A module does not have the signature anti-
parallel ␤-sheet at the N terminus observed in other LDL-A
modules; thus, it has an even less recognizable secondary struc-
ture (28). Comparisons of quail and chicken Tva protein se-
quences indicate that the C1-C2 regions are highly similar
except for two conserved variations and that the C3-C6 regions
are identical or conserved except for one residue. However,
there is significant sequence divergence in the Tva LDL-A
FIG. 6. (A) Constructs of TL5 chimera 2 with substituted C2-C3
regions. C5 has the C2-C3 domain of the chicken Tva LDL-A module,
and L5 has the C2-C3 domain of the LDL-A22 from LRP2. (B) The
abilities of C5 and L5 to mediate ASLV-A infection (as measured
using EnvA-pseudotyped HIV infection). Experiments were repeated
three times with consistent results. Bars, standard deviations. Mock,
mock transfected.
VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 689
module between quail and chicken in the region between the
second and the third cysteines—only four of the nine residues
are conserved (Fig. 7). This sequence divergence may dictate
subtle differences in this region and determine specificity in
receptor usage. Indeed, it was reported that several ASLV-A
variants which were genetically selected for a soluble form of
quail Tva could not use quail Tva but could still efficiently use
chicken Tva as the receptor in viral entry (14–16). These re-
sults seem to suggest that specific residues in the N terminus of
Tva play an important role in specificity in receptor usage.
In contrast, our results have shown that the spacing between
the second and third cysteines also plays a critical role in
receptor function. The C2-C3 regions of both chicken Tva and
LDL-A22 have the same number of residues as the quail Tva
module. However, the individual amino acids are more con-
served between quail and chicken Tva than between quail Tva
and LDL-A22. As mentioned above, four out of nine amino
acids between C2 and C3 are conserved between chicken and
quail Tva proteins, while not a single residue is conserved
between LDL-A22 and quail Tva. Nevertheless, both C5 and
L5 were able to mediate viral entry at a level that was at least
100-fold higher than that seen with TL5LHWG (Fig. 6B).
Since the C2-C3 regions of both LDL-A4 and LDL-A5 have
only four residues (Fig. 7), these results illustrate the impor-
tance of maintaining the proper spacing between C2 and C3 of
Tva for optimal receptor function. These results may appear to
be inconsistent with those of a previous report by Rong et al.
demonstrating that deletion of the C1-C3 region of Tva re-
duced the viral receptor function of Tva by only approximately
50 to 60%. One possible explanation is that the defective
phenotype of the C1-C3 deletion mutant of Tva was masked
due to overexpression of the protein in the previous study (23).
Another possibility is that there are other viral interaction
residues in the C3-C6 region of Tva which are critical for the
viral receptor function but which are not conserved in human
LDL-A5. Thus, the defective phenotype of TL5 LHWG be-
came more apparent than that of the C1-C3 deletion mutant of
Tva in mediation of ASLV-A infection.
In conclusion, the results of the present study have demon-
strated that the spacing between cysteines 2 and 3 of the Tva
LDL-A module plays a crucial role in mediation of ASLV-A
entry. We hypothesize that this region can help maintain the
proper conformation of Tva upon EnvA binding, thus optimiz-
ing Tva-EnvA interactions in ASLV-A entry. It is important to
point out that the results reported here do not exclude the
possibility that some individual residues in this region play a
role in viral entry. We have shown that C5 is approximately
10-fold more efficient than L5 in mediation of viral entry (Fig.
6B), suggesting that certain residues of chicken and quail Tva
in this region are directly involved in ligand contact. Therefore,
the N-terminal region of the Tva LDL-A module is responsible
for both correct protein conformation and ligand recognition
for optimal receptor function in mediations of ASLV-A entry.
ACKNOWLEDGMENTS
We thank Judith White, University of Virginia, for providing the
polyclonal anti-EnvA tail antibody and Gary Cohen and Roselyn
Eisenberg, University of Pennsylvania, for providing the monoclonal
anti-gD antibody. We thank Osvaldo Martinez, University of Illinois at
Chicago, for help in flow cytometry and Osvaldo Martinez and Balaji
Manicassamy for critical reading of the manuscript. The following
reagent was obtained through the AIDS Research and Reference
Reagent Program, Division of AIDS, NIAID NIH: pNL4-3.Luc.R-E
(from Nathaniel Landau).
The research work was supported by American Heart Association
Midwest Affiliate grant-in-aid 9951134Z and National Institutes of
Health grant CA 092459. L.R. was a recipient of the Schweppe Foun-
dation Career Development Award. K.R. was supported by a predoc-
toral fellowship from American Heart Association Midwest Affiliate.
REFERENCES
1. Bates, P., J. A. T. Young, and H. E. Varmus. 1993. A receptor for subgroup
A Rous sarcoma virus is related to the low density lipoprotein receptor. Cell
74:1043–1051.
2. Belanger, C., K. Zingler, and J. A. T. Young. 1995. Importance of cysteines
in the LDLR-related domains of the subgroup A avian leukosis and sarcoma
virus receptor for viral entry. J. Virol. 69:1019–1024.
3. Connolly, L., K. Zingler, and J. A. T. Young. 1994. A soluble form of a
receptor for subgroup A avian leukosis and sarcoma virus (ALSV-A) blocks
infection and binds directly to ALSV-A. J. Virol. 68:2760–2764.
4. Connor, R. I., B. K. Chen, S. Choe, and N. R. Landau. 1995. Vpr is required
for efficient replication of human immunodeficiency virus type-1 in mono-
nuclear phagocytes. Virology 206:935–944.
5. Daly, N. L., J. T. Djordjevic, P. A. Kroon, and R. Smith. 1995. Three-
dimensional structure of the second cysteine-rich repeat from the human
low-density lipoprotein receptor. Biochemistry 34:14474–14481.
6. Daly, N. L., M. J. Scanlon, J. T. Tjordjevic, P. A. Kroon, and R. Smith. 1995.
Three-dimensional structure of a cysteine-rich repeat from the low-density
lipoprotein receptor. Proc. Natl. Acad. Sci. USA 92:6334–6338.
7. Damico, R. L., J. Crane, and P. Bates. 1998. Receptor-triggered membrane
association of a model retroviral glycoprotein. Proc. Natl. Acad. Sci. USA
95:2580–2585.
8. Dolmer, K., W. Huang, and P. G. Gettins. 2000. NMR solution structure of
complement-like repeat CR3 from the low density lipoprotein receptor-
related protein. Evidence for specific binding to the receptor binding domain
of human ␣(2)-macroglobulin. J. Biol. Chem. 275:3264–3269.
9. Fass, D., S. Blacklow, P. S. Kim, and J. M. Berger. 1997. Molecular basis of
familial hypercholesterolaemia from structure of LDL receptor module.
Nature 388:691–693.
10. Federspiel, M. J., P. Bates, J. A. Young, H. E. Varmus, and S. H. Hughes.
1994. A system for tissue-specific gene targeting: transgenic mice susceptible
to subgroup A avian leukosis virus-based retroviral vectors. Proc. Natl. Acad.
Sci. USA 91:11241–11245.
11. Gilbert, J. M., L. D. Hernandez, J. W. Balliet, P. Bates, and J. M. White.
1995. Receptor-induced conformational changes in the subgroup A avian
leukosis and sarcoma virus envelope glycoprotein. J. Virol. 69:7410–7415.
12. He, J., S. Choe, R. Walker, P. Di Marzio, D. O. Morgan, and N. R. Landau.
1995. Human immunodeficiency virus type 1 viral protein R (Vpr) arrests
cells in the G2 phase of the cell cycle by inhibiting p34cdc2 activity. J. Virol.
69:6705–6711.
13. Hernandez, L. D., R. J. Peters, S. E. Delos, J. A. T. Young, D. A. Agard, and
J. M. White. 1997. Activation of a retroviral membrane fusion protein:
soluble receptor-induced liposome binding of the ALSV envelope glycopro-
tein. J. Cell Biol. 139:1455–1464.
14. Holmen, S. L., and M. J. Federspiel. 2000. Selection of a subgroup A avian
leukosis virus [ALV(A)] envelope resistant to soluble ALV(A) surface gly-
coprotein. Virology 273:364–373.
15. Holmen, S. L., D. C. Melder, and M. J. Federspiel. 2001. Identification of key
residues in subgroup A avian leukosis virus envelope determining receptor
binding affinity and infectivity of cells expressing chicken or quail Tva re-
ceptor. J. Virol. 75:726–737.
FIG. 7. Sequence comparison of the C1-C3 regions among the per-
tinent LDL-A modules in this study. Residues are numbered at the top
according to the mature Tva. Residues identical to those of quail Tva
are boxed.
690 RAI ET AL. J. VIROL.
16. Holmen, S. L., D. W. Salter, W. S. Payne, J. B. Dodgson, S. H. Hughes, and
M. J. Federspiel. 1999. Soluble forms of the subgroup A avian leukosis virus
[ALV(A)] receptor Tva significantly inhibit ALV(A) infection in vitro and in
vivo. J. Virol. 73:10051–10060.
17. Huang, W., K. Dolmer, and P. G. Gettins. 1999. NMR solution structure of
complement-like repeat CR8 from the low density lipoprotein receptor-
related protein. J. Biol. Chem. 274:14130–14136.
18. Kozarsky, K., M. Grossman, and J. M. Wilson. 1993. Adenovirus-mediated
correction of the genetic defect in hepatocytes from patients with familial
hypercholesterolemia. Somat. Cell Mol. Genet. 19:449–458.
19. Mothes, W., A. L. Boerger, S. Narayan, J. M. Cunningham, and J. A. T.
Young. 2000. Retroviral entry mediated by receptor priming and low pH
triggering of an envelope glycoprotein. Cell 103:679–689.
20. North, C. L., and S. C. Blacklow. 2000. Solution structure of the sixth LDL-A
module of the LDL receptor. Biochemistry 39:2564–2571.
21. Rong, L., and P. Bates. 1995. Analysis of the subgroup A avian sarcoma and
leukosis virus receptor: the 40-residue, cysteine-rich, low-density lipoprotein
receptor repeat motif of Tva is sufficient to mediate viral entry. J. Virol.
69:4847–4853.
22. Rong, L., K. Gendron, and P. Bates. 1998. Conversion of a human low-
density lipoprotein receptor ligand-binding repeat to a virus receptor: iden-
tification of residues important for ligand specificity. Proc. Natl. Acad. Sci.
USA 95:8467–8472.
23. Rong, L., K. Gendron, B. Strohl, R. Shenoy, R. J. Wool-Lewis, and P. Bates.
1998. Characterization of determinants for envelope binding and infection in
Tva, the subgroup A avian sarcoma and leukosis virus receptor. J. Virol.
72:4552–4559.
24. Rudenko, G., L. Henry, K. Henderson, K. Ichtchenko, M. S. Brown, J. L.
Goldstein, and J. Deisenhofer. 2002. Structure of the LDL receptor extra-
cellular domain at endosomal pH. Science 298:2353–2358.
25. Simonovic, M., K. Dolmer, W. Huang, D. K. Strickland, K. Volz, and P. G.
Gettins. 2001. Calcium coordination and pH dependence of the calcium
affinity of ligand-binding repeat CR7 from the LRP. Comparison with re-
lated domains from the LRP and the LDL receptor. Biochemistry 40:15127–
15134.
26. Tonelli, M., R. J. Peters, T. L. James, and D. A. Agard. 2001. The solution
structure of the viral binding domain of Tva, the cellular receptor for sub-
group A avian leukosis and sarcoma virus. FEBS Lett. 509:161–168.
27. Wang, Q. Y., K. Dolmer, W. Huang, P. G. Gettins, and L. Rong. 2001. Role
of calcium in protein folding and function of Tva, the receptor of subgroup
A avian sarcoma and leukosis virus. J. Virol. 75:2051–2058.
28. Wang, Q. Y., W. Huang, K. Dolmer, P. G. Gettins, and L. Rong. 2002.
Solution structure of the viral receptor domain of Tva and its implications in
viral entry. J. Virol. 76:2848–2856.
29. Young, J. A. T., P. Bates, and H. E. Varmus. 1993. Isolation of a chicken gene
that confers susceptibility to infection by subgroup A avian leukosis and
sarcoma viruses. J. Virol. 67:1811–1816.
30. Yu, X., Q. Y. Wang, Y. Guo, K. Dolmer, J. A. Young, P. G. Gettins, and L.
Rong. 2003. Kinetic analysis of binding interaction between the subgroup A
Rous sarcoma virus glycoprotein SU and its cognate receptor Tva: calcium is
not required for ligand binding. J. Virol. 77:7517–7526.
31. Zingler, K., and J. A. T. Young. 1996. Residue Trp-48 of Tva is critical for
viral entry but not for high-affinity binding to the SU glycoprotein of sub-
group A avian leukosis and sarcoma viruses. J. Virol. 70:7510–7516.
32. Zingler, K., C. Belanger, R. Peters, D. Agard, and J. A. T. Young. 1995.
Identification and characterization of the viral interaction determinants of
the subgroup A avian leukosis virus receptor. J. Virol. 69:4261–4266.
VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 691

More Related Content

What's hot

Directed evolution of a recombinase that excises the provirus of most HIV-1 p...
Directed evolution of a recombinase that excises the provirus of most HIV-1 p...Directed evolution of a recombinase that excises the provirus of most HIV-1 p...
Directed evolution of a recombinase that excises the provirus of most HIV-1 p...Carola Schäfer
 
NAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studiesNAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studiesShih-hsin Kan
 
Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...
Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...
Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...gan-navi
 
The Thiazide-sensitive NaCl
The Thiazide-sensitive NaClThe Thiazide-sensitive NaCl
The Thiazide-sensitive NaClAvin Snyder
 
A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...
A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...
A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...Igor Putrenko
 
project- Liron Schwartz
project- Liron Schwartzproject- Liron Schwartz
project- Liron SchwartzLiron Schwartz
 
Activation of surrogate death receptor signalling triggers peroxynitrite depe...
Activation of surrogate death receptor signalling triggers peroxynitrite depe...Activation of surrogate death receptor signalling triggers peroxynitrite depe...
Activation of surrogate death receptor signalling triggers peroxynitrite depe...Saurabh Shekhar
 
Phosphoproteomics of collagen receptor networks reveals SHP-2
Phosphoproteomics of collagen receptor networks reveals SHP-2Phosphoproteomics of collagen receptor networks reveals SHP-2
Phosphoproteomics of collagen receptor networks reveals SHP-2Maciej Luczynski
 
Irvin PLoS Genet 2014
Irvin PLoS Genet 2014Irvin PLoS Genet 2014
Irvin PLoS Genet 2014Jordan Irvin
 
Dimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi and
Dimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi andDimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi and
Dimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi andrkkoiri
 
FabioAmaralProject 3
FabioAmaralProject 3FabioAmaralProject 3
FabioAmaralProject 3Fabio Amaral
 
Catterall molecular mechanism of calcium channel regulation in the
Catterall molecular mechanism of calcium channel regulation in theCatterall molecular mechanism of calcium channel regulation in the
Catterall molecular mechanism of calcium channel regulation in theramezali
 

What's hot (20)

nature
naturenature
nature
 
Directed evolution of a recombinase that excises the provirus of most HIV-1 p...
Directed evolution of a recombinase that excises the provirus of most HIV-1 p...Directed evolution of a recombinase that excises the provirus of most HIV-1 p...
Directed evolution of a recombinase that excises the provirus of most HIV-1 p...
 
NAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studiesNAGLU_IGF2 in vitro studies
NAGLU_IGF2 in vitro studies
 
Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...
Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...
Cdc6 Knockdown Renders p16INK4a Re-Activation, Leading to Senescence Human Br...
 
The Thiazide-sensitive NaCl
The Thiazide-sensitive NaClThe Thiazide-sensitive NaCl
The Thiazide-sensitive NaCl
 
A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...
A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...
A family of acetylcholine-gated chloride channel subunits in Caenorhabditis e...
 
1135-05
1135-051135-05
1135-05
 
project- Liron Schwartz
project- Liron Schwartzproject- Liron Schwartz
project- Liron Schwartz
 
Activation of surrogate death receptor signalling triggers peroxynitrite depe...
Activation of surrogate death receptor signalling triggers peroxynitrite depe...Activation of surrogate death receptor signalling triggers peroxynitrite depe...
Activation of surrogate death receptor signalling triggers peroxynitrite depe...
 
Phosphoproteomics of collagen receptor networks reveals SHP-2
Phosphoproteomics of collagen receptor networks reveals SHP-2Phosphoproteomics of collagen receptor networks reveals SHP-2
Phosphoproteomics of collagen receptor networks reveals SHP-2
 
JBC2
JBC2JBC2
JBC2
 
2009 coumarin aaa induces apoptosis like cell death
2009 coumarin aaa induces apoptosis like cell death2009 coumarin aaa induces apoptosis like cell death
2009 coumarin aaa induces apoptosis like cell death
 
Irvin PLoS Genet 2014
Irvin PLoS Genet 2014Irvin PLoS Genet 2014
Irvin PLoS Genet 2014
 
Dimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi and
Dimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi andDimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi and
Dimethyl sulfoxide activates tumorn ecrosisfactor p53 mediated apoptosi and
 
FabioAmaralProject 3
FabioAmaralProject 3FabioAmaralProject 3
FabioAmaralProject 3
 
ProteinSci2007
ProteinSci2007ProteinSci2007
ProteinSci2007
 
Catterall molecular mechanism of calcium channel regulation in the
Catterall molecular mechanism of calcium channel regulation in theCatterall molecular mechanism of calcium channel regulation in the
Catterall molecular mechanism of calcium channel regulation in the
 
Wp seidman 12
Wp seidman 12Wp seidman 12
Wp seidman 12
 
Poster_mainFin1
Poster_mainFin1Poster_mainFin1
Poster_mainFin1
 
JClinChem_2003
JClinChem_2003JClinChem_2003
JClinChem_2003
 

Viewers also liked

African Standby Force media toolkit
African Standby Force media toolkit African Standby Force media toolkit
African Standby Force media toolkit Jacqueline Cochrane
 
Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...
Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...
Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...Małgorzata Sikora-Gaca
 
Confiture et concentré de tomates
Confiture et concentré de tomatesConfiture et concentré de tomates
Confiture et concentré de tomatesZiad Yassine
 
Hamilton Youth Engagement Initiative : Year in Review 2012-2013
Hamilton Youth Engagement Initiative : Year in Review 2012-2013Hamilton Youth Engagement Initiative : Year in Review 2012-2013
Hamilton Youth Engagement Initiative : Year in Review 2012-2013Alex Ramirez
 
CEREBRO TRIUNO
CEREBRO TRIUNOCEREBRO TRIUNO
CEREBRO TRIUNOPaul Lobo
 
Eumind about_me_ppt___water_footprint
Eumind  about_me_ppt___water_footprintEumind  about_me_ppt___water_footprint
Eumind about_me_ppt___water_footprintsarthakgarg123
 
Oge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsya
Oge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsyaOge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsya
Oge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsyaЕлена Павлова
 
Ms.Powerpoint BAB 5
Ms.Powerpoint BAB 5Ms.Powerpoint BAB 5
Ms.Powerpoint BAB 5Zahra Rizqi
 
Continguts del primer trimestre curs 2015
Continguts del primer trimestre curs 2015Continguts del primer trimestre curs 2015
Continguts del primer trimestre curs 2015Martalfonsog
 
online assignment
online assignmentonline assignment
online assignmentsivapriyags
 
Pregnancy- Human Development
Pregnancy- Human Development Pregnancy- Human Development
Pregnancy- Human Development jchartrand97
 

Viewers also liked (17)

African Standby Force media toolkit
African Standby Force media toolkit African Standby Force media toolkit
African Standby Force media toolkit
 
Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...
Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...
Perspektywa finansowa 2014-2020. Charakterystyka Programu Operacyjnego Inteli...
 
Confiture et concentré de tomates
Confiture et concentré de tomatesConfiture et concentré de tomates
Confiture et concentré de tomates
 
Hamilton Youth Engagement Initiative : Year in Review 2012-2013
Hamilton Youth Engagement Initiative : Year in Review 2012-2013Hamilton Youth Engagement Initiative : Year in Review 2012-2013
Hamilton Youth Engagement Initiative : Year in Review 2012-2013
 
Dine
DineDine
Dine
 
CEREBRO TRIUNO
CEREBRO TRIUNOCEREBRO TRIUNO
CEREBRO TRIUNO
 
Parallel Plates
Parallel PlatesParallel Plates
Parallel Plates
 
Eumind about_me_ppt___water_footprint
Eumind  about_me_ppt___water_footprintEumind  about_me_ppt___water_footprint
Eumind about_me_ppt___water_footprint
 
Oge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsya
Oge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsyaOge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsya
Oge 2016 literatura_komplex_materialov_dlya_podgotovki_uchaschikhsya
 
Ms.Powerpoint BAB 5
Ms.Powerpoint BAB 5Ms.Powerpoint BAB 5
Ms.Powerpoint BAB 5
 
Cardillo rocco
Cardillo roccoCardillo rocco
Cardillo rocco
 
Continguts del primer trimestre curs 2015
Continguts del primer trimestre curs 2015Continguts del primer trimestre curs 2015
Continguts del primer trimestre curs 2015
 
online assignment
online assignmentonline assignment
online assignment
 
Pregnancy- Human Development
Pregnancy- Human Development Pregnancy- Human Development
Pregnancy- Human Development
 
FIREFOX OS...
FIREFOX OS...FIREFOX OS...
FIREFOX OS...
 
Giuseffi graziano
Giuseffi grazianoGiuseffi graziano
Giuseffi graziano
 
Seminar 1 scientists
Seminar 1 scientistsSeminar 1 scientists
Seminar 1 scientists
 

Similar to 1429

BOMCl etherial nature of AGPS
BOMCl etherial nature of AGPSBOMCl etherial nature of AGPS
BOMCl etherial nature of AGPSKendal Ryter
 
Alignment of polysaccharide sequences
Alignment of polysaccharide sequencesAlignment of polysaccharide sequences
Alignment of polysaccharide sequencesRoland Stenutz
 
Expression, purification and spectroscopic characterization of the cytochrome...
Expression, purification and spectroscopic characterization of the cytochrome...Expression, purification and spectroscopic characterization of the cytochrome...
Expression, purification and spectroscopic characterization of the cytochrome...John Clarkson
 
2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...
2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...
2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...Simon Gemble
 
Sequencing the Human TCRβ Repertoire on the Ion S5TM
Sequencing the Human TCRβ Repertoire on the Ion S5TMSequencing the Human TCRβ Repertoire on the Ion S5TM
Sequencing the Human TCRβ Repertoire on the Ion S5TMThermo Fisher Scientific
 
IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...
IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...
IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...IRJET Journal
 
Viroids and the RNA World
Viroids and the RNA WorldViroids and the RNA World
Viroids and the RNA WorldFabrice Leclerc
 
3aff_monooxygensae.pdf
3aff_monooxygensae.pdf3aff_monooxygensae.pdf
3aff_monooxygensae.pdfssuserc7b94d
 
Structural Determinants of Drugs Acting on the Nav1.8 Channel
Structural Determinants of Drugs Acting on the Nav1.8 ChannelStructural Determinants of Drugs Acting on the Nav1.8 Channel
Structural Determinants of Drugs Acting on the Nav1.8 ChannelShahnaz Yusaf
 
Rosloniec_et_al-2009-Molecular_Microbiology
Rosloniec_et_al-2009-Molecular_MicrobiologyRosloniec_et_al-2009-Molecular_Microbiology
Rosloniec_et_al-2009-Molecular_MicrobiologyKamila du Plessis
 
CAPE Biology 2015 Unit 1 paper 1
CAPE Biology 2015 Unit 1 paper 1 CAPE Biology 2015 Unit 1 paper 1
CAPE Biology 2015 Unit 1 paper 1 CAPEStudentHelp
 
T CELL RECEPTOR.pptx
T CELL RECEPTOR.pptxT CELL RECEPTOR.pptx
T CELL RECEPTOR.pptxBinteHawah1
 
Chan et al 2013 abasic site bypass
Chan et al 2013 abasic site bypassChan et al 2013 abasic site bypass
Chan et al 2013 abasic site bypassKin Chan
 

Similar to 1429 (20)

BOMCl etherial nature of AGPS
BOMCl etherial nature of AGPSBOMCl etherial nature of AGPS
BOMCl etherial nature of AGPS
 
Alignment of polysaccharide sequences
Alignment of polysaccharide sequencesAlignment of polysaccharide sequences
Alignment of polysaccharide sequences
 
Expression, purification and spectroscopic characterization of the cytochrome...
Expression, purification and spectroscopic characterization of the cytochrome...Expression, purification and spectroscopic characterization of the cytochrome...
Expression, purification and spectroscopic characterization of the cytochrome...
 
2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...
2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...
2015 - Cdk5 promotes DNA replication stress checkpoint activation through RPA...
 
Sequencing the Human TCRβ Repertoire on the Ion S5TM
Sequencing the Human TCRβ Repertoire on the Ion S5TMSequencing the Human TCRβ Repertoire on the Ion S5TM
Sequencing the Human TCRβ Repertoire on the Ion S5TM
 
IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...
IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...
IRJET- Silencing of hnRNP A1 and hnRNP A2/B1 Downregulates the Expression of ...
 
Viroids and the RNA World
Viroids and the RNA WorldViroids and the RNA World
Viroids and the RNA World
 
Celis 2015, KpCld
Celis 2015, KpCldCelis 2015, KpCld
Celis 2015, KpCld
 
3aff_monooxygensae.pdf
3aff_monooxygensae.pdf3aff_monooxygensae.pdf
3aff_monooxygensae.pdf
 
Blaiser poster
Blaiser posterBlaiser poster
Blaiser poster
 
IJBM2014
IJBM2014IJBM2014
IJBM2014
 
Dako handbook
Dako handbookDako handbook
Dako handbook
 
Structural Determinants of Drugs Acting on the Nav1.8 Channel
Structural Determinants of Drugs Acting on the Nav1.8 ChannelStructural Determinants of Drugs Acting on the Nav1.8 Channel
Structural Determinants of Drugs Acting on the Nav1.8 Channel
 
Session 1.2: Chiorazzi
Session 1.2: ChiorazziSession 1.2: Chiorazzi
Session 1.2: Chiorazzi
 
Session 1.2 Chiorazzi
Session 1.2 ChiorazziSession 1.2 Chiorazzi
Session 1.2 Chiorazzi
 
Rosloniec_et_al-2009-Molecular_Microbiology
Rosloniec_et_al-2009-Molecular_MicrobiologyRosloniec_et_al-2009-Molecular_Microbiology
Rosloniec_et_al-2009-Molecular_Microbiology
 
CAPE Biology 2015 Unit 1 paper 1
CAPE Biology 2015 Unit 1 paper 1 CAPE Biology 2015 Unit 1 paper 1
CAPE Biology 2015 Unit 1 paper 1
 
T CELL RECEPTOR.pptx
T CELL RECEPTOR.pptxT CELL RECEPTOR.pptx
T CELL RECEPTOR.pptx
 
Chan et al 2013 abasic site bypass
Chan et al 2013 abasic site bypassChan et al 2013 abasic site bypass
Chan et al 2013 abasic site bypass
 
Nagabhushana_poster
Nagabhushana_posterNagabhushana_poster
Nagabhushana_poster
 

1429

  • 1. JOURNAL OF VIROLOGY, Jan. 2004, p. 683–691 Vol. 78, No. 2 0022-538X/04/$08.00ϩ0 DOI: 10.1128/JVI.78.2.683–691.2004 Copyright © 2004, American Society for Microbiology. All Rights Reserved. The Spacing between Cysteines Two and Three of the LDL-A Module of Tva Is Important for Subgroup A Avian Sarcoma and Leukosis Virus Entry Tia Rai, Deborah Marble, Kayla Rihani, and Lijun Rong* Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612 Received 17 July 2003/Accepted 3 October 2003 Rong et al. have demonstrated previously that with a few substitutions, the fourth repeat of human low-density lipoprotein (hLDL-A4) receptor can functionally replace the LDL-A module of Tva, the cellular receptor for subgroup A avian sarcoma and leukosis virus (ASLV-A), in viral entry (L. Rong, K. Gendron, and P. Bates, Proc. Natl. Acad. Sci. USA 95:8467-8472, 1998). Here we have shown that swapping the amino terminus of hLDL repeat 5 (hLDL-A5) with that of Tva, in addition to the corresponding substitutions made in human LDL-A4, was required to convert hLDL-A5 into an efficient ASLV-A receptor. These results sub- stantiated our previous findings regarding the role of the specific residues in the viral interaction domain of Tva and demonstrated the critical role of the amino terminus of the Tva LDL-A module in ASLV-A infection. Furthermore, we have shown that the residues between cysteines 2 and 3 of the Tva LDL-A module in a Tva/LDL-A5 chimeric protein can be functionally replaced by the corresponding region of another LDL-A module, human LDL receptor-related protein repeat 22 (LDL-A22), to mediate efficient ASLV-A entry. Since the only conserved feature between the C2-C3 region of LDL-A22 and the Tva LDL-A module is that both contain nine amino acids of which none are conserved, we conclude that the spacing between C2 and C3 of the LDL-A module of Tva is an important determinant for ASLV-A entry. Thus, the present study provides strong evidence to support our hypothesis that one role of the N terminus of the LDL-A module of Tva is to allow proper folding and conformation of the protein for optimal interaction with the viral glycoprotein EnvA in ASLV-A entry. Entry of subgroup A avian sarcoma and leukosis virus (ASLV-A) into its hosts is mediated by interactions between the viral glycoprotein EnvA and a small cell surface glycopro- tein, Tva (1, 29). Direct interaction of EnvA with Tva is not only important for initial viral attachment but is also important for the postbinding steps in ASLV-A entry. Binding between Tva and EnvA in vitro can induce a series of conformational changes on EnvA that lead to exposure of the putative fusion peptide and its association with the lipid membrane (7, 11, 13, 19). These results provide direct biochemical evidence that receptor binding can trigger the structural rearrangements likely required for membrane fusion mediated by a viral gly- coprotein. Since receptor triggering appears to be a common yet poorly understood mechanism for entry of various envel- oped viruses, analysis of the Tva-EnvA interaction serves as an excellent model in elucidating the basic principles in viral en- try. One of the major advantages of using Tva/EnvA as a model to elucidate the viral entry mechanism is that the viral inter- action domain of Tva is solely determined by a single low- density lipoprotein (LDL) receptor module within the extra- cellular domain of Tva (3, 21), making it a simple model amenable to molecular, biochemical, and structural analysis. The LDL-A module of Tva is 40 amino acids in length and includes six invariable cysteines and five highly conserved acidic residues found in other LDL-A modules such as human LDL receptor and LDL receptor-related proteins (1). Bio- chemical and structural analysis of different LDL-A modules demonstrates that the six conserved cysteines are involved in the formation of three pairs of disulfide bonds. Furthermore, structural analysis of several individual LDL-A modules (and, recently, of the entire ectodomain of human LDL receptor) by X-ray crystallography showed that the side chains of four con- served acidic residues near the C terminus and the carbonyl oxygen groups of two nonacidic residues of each LDL-A mod- ule coordinate calcium binding (9, 24, 25). These common structural features of LDL-A modules are important for proper folding and, thus, for their functions in ligand binding. The role of the Tva LDL-A module in EnvA binding and ASLV-A entry has been extensively examined by molecular, biochemical, and structural analysis. It has been demonstrated that the LDL-A module of Tva efficiently mediated ASLV-A entry when it was appended to a heterologous membrane- spanning domain (21). Furthermore, this module of Tva can be functionally replaced by a modified human LDL-A4 module in mediation of ASLV-A entry (22). These studies, together with mutational analysis (23, 31, 32), have identified several puta- tive viral interaction residues important for viral entry. Bio- chemical and structural analysis demonstrated that like other LDL-A modules, the correct in vitro folding of the Tva LDL-A module is calcium dependent (26–28). Surprisingly, however, calcium is not essential for EnvA binding once the protein is correctly folded (30). * Corresponding author. Mailing address: Department of Microbi- ology and Immunology, College of Medicine, University of Illinois at Chicago, E829 MSB, 835 S. Wolcott Ave., Chicago, IL 60612. Phone: (312) 355-0203. Fax: (312) 996-6415. E-mail: lijun@uic.edu. 683
  • 2. One important structural difference between the Tva and other LDL-A modules is that the Tva LDL-A module does not have the signature anti-parallel ␤-sheet observed at the N-terminal C1-C3 region of other LDL-A modules (5, 6, 8, 9, 17, 20, 25, 26, 28). The C2-C3 region of the Tva LDL-A module contains nine residues instead of the four to five residues seen in the other LDL-A modules, and this region of Tva is responsible for the flexible conformation at the N terminus (28). In this study, we wanted to examine the role of this region in determining the receptor function of Tva. We found that the spacing between the C2 and C3 regions of the Tva LDL-A module is an important determinant for ASLV-A entry. The present study provides strong evidence that one role of the N-terminal region of the Tva LDL-A module is to allow proper folding and overall conformation of the protein for optimal interaction with EnvA in ASLV-A entry. MATERIALS AND METHODS Cells and viruses. Human embryonic kidney 293T cells were maintained as described previously (22). RCAS(A)AP (10) viral stocks were generated as described previously (21). The EnvA-pseudotyped human immunodeficiency virus (HIV) viral stocks were generated using the HIV vector pNL4-3.Luc.R-E (4, 12) cotransfected with ASLV-A glycoprotein EnvA into 293T cells. Briefly, 293T cells were transiently transfected with 10 ␮g of the HIV vector either alone or with 10 ␮g of EnvA DNA or vesicular stomatitis virus glycoprotein G (VSV-G) DNA by a modified CaPO4 method as previously described (21). The supernatants (viral stocks) were harvested 48 h posttransfection, filtered through a 0.22-␮m-pore-size filter, and stored at Ϫ80°C. Cloning, mutagenesis, and mutant nomenclature. A chimeric Tva construct containing the human LDL-A5 instead of the Tva LDL-A module was generated as follows. The PCR-amplified coding region for human LDL-A5 was digested (using human LDL receptor cDNA as the template) (18) with restriction endo- nucleases BamHI and SacII, and the digested DNA fragment was inserted into the BamHI/SacII-digested Myc-Tva vector (22). This construct is referred to as TL5, where T stands for Tva and L5 pertains to the LDL-A5 region. The other TL5 derivatives were generated using the TL5 construct as the template by a standard two-step PCR protocol. Four amino acids in the TL5 construct, namely, FIG. 1. (A) Sequence alignment of quail Tva LDL-A module with human LDL-A4 and LDL-A5 receptors 4 and 5. Residues are numbered according to the sequence of the mature Tva (1). The six invariable cysteines are boxed. The consensus amino acids among these and other LDL-A modules are shown at the bottom. (B) Tva/hLDL-A5 constructs generated in this study. Human LDL-A5 (hLDL-A5) is 35 amino acids in length and is numbered from 1 to 35. The four residues in LDL-A5 that were targeted are underlined. The nomenclature of the TL5 constructs is similar to that used previously for the TL4 constructs. 684 RAI ET AL. J. VIROL.
  • 3. R19, G23, E33, and G34, were replaced with the corresponding residues of the Tva LDL-A module either individually or in combination (Fig. 1B). Two addi- tional TL5 chimeric constructs were generated. TL5 chimera 1 has the N- terminal C1-C3 region from the Tva LDL-A module and also contains the substitutions R19L, G23H, and E33W. TL5 chimera 2 is same as TL5 chimera 1 except that it also has the N34G substitution (Fig. 1B). In addition, constructs C5 and L5 were generated using TL5 chimera 2 as the template but contained the C2-C3 region from the chicken Tva LDL-A module and the C2-C3 region of the human LDL-A22 module of LRP2, respectively. Analysis of protein expression. Human kidney embryonic 293T cells were transiently transfected with Myc-Tva and the TL5 constructs by a modified CaPO4 method. Cells were lysed 48 h after transfection, and protein expression was examined by Western blotting as previously described (22). Myc-tagged Tva and TL5 proteins were detected using anti-myc monoclonal antibody (MAb) 9E10 as the primary antibody. Protein surface expression was examined by fluorescence-activated cell sorter (FACS) analysis. Briefly, 293T cells were transiently transfected with 10 ␮g of DNAs of myc-Tva and TL5 constructs by the CaPO4 method. At 48 h posttrans- fection, cells were resuspended in FACS buffer (phosphate-buffered saline con- taining 2% fetal calf serum) to 5 ϫ 106 cells/ml. Cells were seeded into a U-bottom 96-well plate and were incubated with 50 ␮l of mouse MAb 9E10 (10 ␮g/ml stock) for 30 min. Cells were then washed twice with FACS buffer and incubated with 50 ␮l of fluorescein isothiocyanate-conjugated rat anti-mouse immunoglobulin G1 antibody at a final concentration of 10 ␮g/ml for 20 min. Cells were washed again and resuspended in 300 ␮l of FACS buffer. Cells were stained for viability with 0.5 ␮l of propidium iodide and were analyzed using a Becton Dickinson FACSCalibur flow cytometer and CellQuest software. ELISA-based binding test. To examine the ability of the TL5 chimeric proteins to bind EnvA, an enzyme-linked immunosorbent assay (ELISA)-based binding test, which was modified from a published protocol (22, 23), was developed. Human 293T cells were transiently transfected with gD-EnvA, myc-Tva, and the TL5 chimeric constructs shown in Fig. 1B. The transfected cells were induced with 10 mM sodium butyrate 24 h posttransfection and lysed 40 h posttransfec- tion with 1% Triton lysis buffer. Protein expression was examined and semiquan- tified by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS- PAGE) and Western blotting using MAbs 9E10 (for myc-tagged Tva and TL5 constructs) and 1D3 (for gD-tagged EnvA). The ELISA binding test was per- formed as follows: 96-well plates were coated with MAb 9E10 for 24 h. After three washings, the plates were blocked for 30 min with phosphate-buffered saline containing 0.05% Tween 20, 0.5% bovine serum albumin, and 0.05% sodium azide. A total of 100 ml of lysates from cells transfected with wild-type (wt) Tva or TL5 construct DNAs was added to each well for 1 h with shaking at 4°C. The plates were then washed three times with distilled water, and 100 ␮l of gD-EnvA lysate was added to each well for 1 h with shaking. The wells were washed and incubated with an anti-EnvA tail rabbit antibody for one more hour at 4°C. Finally, the plates were washed four times, ABTS [2,2Ј-azinobis (3- ethylbenzthiazolinesulfonic acid] and hydrogen peroxide were added to each well, and the absorbance at 405 nm was read after incubation. Each experiment was performed in triplicate, and the relative ability of each protein to bind EnvA was expressed as the percentage of wt Tva binding to EnvA. Infectivity assays. Two different viral systems were employed in this study: a recombinant ASLV-A, RCAS(A)AP, which carries an alkaline phosphatase (AP) gene as the reporter (10), and the EnvA-pseudotyped HIV viruses, which carry a luciferase gene as the reporter (4, 12). The infectivity assay was carried out as previously described (21). Briefly, 293T cells were transiently transfected with DNAs of the Tva or TL5 constructs by the CaPO4 method. At 24 h posttransfection, cells were seeded in six-well plates and then challenged with RCAS(A)AP- or the EnvA-pseudotyped HIV viruses. The RCAS(A)AP-in- fected cells were stained for AP, and the AP-positive cells were enumerated under a microscope. The EnvA-pseudotyped HIV-infected cells were lysed, and luciferase activity was measured with a standard luminometer. RESULTS Human LDL-A5 cannot replace the Tva LDL-A module for viral receptor function. A chimeric Tva protein called TL5, in which the Tva LDL-A module was replaced with human LDL receptor repeat 5 (LDL-A5) (Fig. 1B), was created. To test its ability to mediate viral entry, TL5 DNA was transiently trans- fected into 293T cells and the transfected cells were challenged with a recombinant ASLV-A carrying an AP reporter gene [RCAS(A)AP)]. As expected, TL5 was unable to mediate viral entry (data not shown). To ensure that the defect was not due to a lack of protein expression, the TL5 transfected cells were analyzed by Western blotting; it was found that total protein expression was comparable to that of a myc-tagged wt Tva (Fig. 2, lanes 1 and 2). The surface expression was measured by flow cytometry, and it was found indistinguishable from that of wt Tva (data not shown). In addition, TL5 was also examined, using an ELISA-based test (described later and in Materials and Methods), for its ability to bind the ASLV-A glycoprotein EnvA. As expected, TL5 was unable to bind EnvA (see Fig. 5). These results are similar to the previous findings with TL4, a chimeric protein between Tva and human LDL-A4 which was shown to be unable to bind EnvA and which could not mediate ASLV-A entry (22). Human LDL-A5 with substitutions corresponding to the viral interaction residues of Tva cannot be converted into an effective viral receptor. Previously it was shown that a human LDL-A4 module with a few amino acid substitutions could functionally substitute for the Tva LDL-A module in media- tion of ASLV-A entry (22). This gain-of-function approach was instrumental in identifying several putative viral interaction FIG. 2. Analysis of protein expression of TL5 constructs. Transient expression of the TL5 constructs was examined by Western blotting following SDS-PAGE. At 48 h posttransfection, 293T cells were lysed with Triton lysis buffer and aliquots of samples were subjected to SDS-PAGE and Western blotting. Molecular mass markers (in kilodaltons) are shown to the right of the panel. VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 685
  • 4. residues in Tva. Sequence comparison of human LDL-A5 with human LDL-A4 and the Tva LDL-A module indicated that none of the putative interaction residues of Tva, Leu34, His38, Trp48, and Gly49 are conserved in LDL-A5 (Fig. 1A). To test whether TL5 could be converted into a functional ASLV-A receptor, 11 TL5 substitution variants were generated in which the corresponding residues in TL5 were replaced with the four putative viral interaction residues of Tva either individually or in combination (Fig. 1B). Human 293T cells were transiently transfected with the TL5 construct DNAs and lysed. Protein expression was examined by Western blotting using 9E10, an antibody which specifically recognizes the myc tag portion of these proteins. All 11 TL5 constructs were expressed well in 293T cells (Fig. 2, lanes 3, 4, 5, 7, 8, 9, and 11 to 15). As was previously observed with wt Tva and TL4 constructs (22, 23), each of the TL5 proteins migrated as a smear of multiple bands on SDS-PAGE, indicating that these proteins were heavily modified by glycosylation in 293T cells. To examine the ability of the TL5 proteins to mediate ASLV-A infection, the 293T cells transiently transfected with the TL5 construct DNAs were challenged with the RCAS(A)AP and the recombinant ASLV-A vector; the in- fected cells were enumerated postinfection. Since expression of Tva in native avian cells is extremely low, we were concerned that overexpression of the TL5 constructs in 293T cells by transient transfection might mask the defect of these proteins in mediation of viral entry. To avoid this potential problem, various amounts (1, 5, and 20 ␮g) of DNA of each TL5 con- struct as well as wt Tva were used in transfection prior to RCAS(A)AP challenges. Protein expression was detectable by Western blotting and flow cytometry when 5 or 20 ␮g of DNA was used in transfection (data not shown). As shown in Fig. 3, challenging the wt Tva DNA-transfected 293T cells with RCAS(A)AP viruses resulted in high numbers of AP-positive cells (approximately 105 AP-positive cells per milliliter of viral stock used) when either a low or high amount of DNA was used in transfection, indicating that wt Tva can efficiently mediate ASLV-A entry. As a negative control, mock-transfected 293T cells were also challenged with RCAS(A)AP viruses; no background AP-positive cells were detected. Among the 11 TL5 constructs, only three (TL5LHG, TL5LHW, and TL5LHWG) mediated low levels of ASLV-A infection at a level at least 100-fold lower than that seen with wt Tva (Fig. 3). The rest of the TL5 constructs did not display any viral receptor function (data not shown). These results are in stark contrast with those seen with the TL4 chimeric con- structs with respect to mediation of ASLV-A infection, as previously reported (22). For example, while TL5LHWG was approximately 100-fold lower than that seen with wt Tva, the similar TL4 construct (TL4G-A19LD23H) was able to mediate efficient viral entry. One plausible explanation is that human LDL-A4 is more conserved than human LDL-A5 with respect to the Tva LDL-A module between cysteines 3 and 6 (Fig. 1A). These results suggest that additional viral interaction determi- nants must be critical for maintaining the optimal viral recep- tor function of Tva. The amino terminus of the Tva LDL-A module is required to convert human LDL-A5 into an efficient viral receptor. Previ- ous reports suggest that the amino-terminal region of the Tva LDL-A module plays a subtle role in mediation of ASLV-A infection (22, 23). Thus, we generated two additional chimeric constructs (named TL5 chimera 1 and TL5 chimera 2) which replaced the human LDL-A5 C1-C3 region of TL5LHW and TL5LHWG with the corresponding region of Tva (Fig. 1B). These constructs were expressed well in 293T cells compared to the other TL5 proteins (Fig. 2, lanes 6 and 10). TL5 chimera 1 mediated low levels of ASLV-A infection, while TL5 chimera 2 was as efficient as wt Tva in mediation of viral infection in transfected 293T cells challenged with the RCAS(A)AP vi- FIG. 3. The abilities of TL5 constructs to mediate ASLV-A entry as determined using RCAS(A)AP vector. Human 293T cells transiently transfected with various amounts of plasmid DNA of the constructs shown in Fig. 1 were challenged with a series of 10-fold dilutions of RCAS(A)AP, a recombinant ASLV-A virus carrying an AP reporter gene. Infected cells were fixed with 2% paraformaldehyde at 48 h postinfection and stained for AP activity. The ability of a construct to mediate viral infection is expressed as the number of AP-positive cells (in international units) per milliliter of virus stock used. Experiments were repeated several times with consistent results, and the results of a representative experiment are shown here. 686 RAI ET AL. J. VIROL.
  • 5. ruses (Fig. 3). These results provide strong evidence that the region between cysteines 1 and 3 of the Tva LDL-A module plays a crucial role in the viral receptor function of Tva. In addition, since TL5 chimera 1 and TL5 chimera 2 differ by a single residue (Gln in TL5 chimera 1 versus Gly in TL5 chi- mera 2) (Fig. 1B), these results suggest that maintaining a small residue-like glycine at this position is important for the viral receptor function of the TL5 constructs, a finding fully consistent with the previous report by Rong et al. of investi- gations with human LDL-A4 (22). Examination by an EnvA-pseudotyped HIV infection assay of ASLV-A entry for TL5 constructs. The assay described above, which uses a recombinant ASLV-A virus for infection followed by AP staining and counting under a microscope for infectivity, is a reliable method for measurement and compar- ison of the abilities of Tva and its derivatives or mutants to mediate viral entry. However, one major drawback of this assay is that counting under a microscope is time consuming and error prone. Thus, we used the luciferase gene as the reporter to establish an HIV-based pseudotyping system to quantita- tively measure ASLV-A entry. To test the feasibility and specificity of this system, the DNA of HIV vector pNL4-3.Luc.R-E (4, 12) was cotransfected with the DNAs of either the ASLV-A glycoprotein EnvA or VSV-G to generate the pseudotyped HIV viral stocks (see Materials and Methods for the details). The produced viral stocks were used to challenge mock-transfected human 293T cells or the 293T cells transiently expressing Tva, and the luciferase activity of the cells was determined with a luminometer. The cells infected by the VSV-G-pseudotyped HIV viral stock, as ex- pected, gave a high level of luciferase activity (approximately 8 logs of relative light units [RLU]) regardless of whether Tva was expressed or not (Fig. 4A), indicating that VSV-G medi- ated entry is Tva independent. In contrast, the cells challenged with the EnvA-pseudotyped HIV viruses only gave a high level of luciferase activity (also about 8 logs of RLU) when the cells expressed Tva. However, the cells challenged by the HIV viri- ons lacking a glycoprotein, or the cells without Tva expression challenged by the EnvA-pseudotyped HIV virions, only gave background luciferase activity (Fig. 4A). These results indicate that EnvA-mediated HIV entry is Tva dependent, as expected, demonstrating the feasibility and specificity of the EnvA- pseudotyped HIV system to measure ASLV-A entry. The abilities of the TL5 chimeric constructs (shown in Fig. 3) to mediate ASLV-A entry were also tested using the EnvA- pseudotyped HIV viruses. TL5 chimera 2 could mediate ASLV-A entry as efficiently as wt Tva, while TL5 chimera 1 was at least three logs less efficient than Tva. Furthermore, TL5LHWG was better than TL5LHW, while TL5LHG was unable to mediate ASLV-A entry (Fig. 4B). Thus, the results were remarkably consistent with that seen with RCAS(A)AP infection (Fig. 3). Therefore, we have established an easy and quantitative assay to measure ASLV-A entry; this assay will facilitate studies of Tva-EnvA interactions. Most TL5 proteins do not efficiently bind to ASLV-A glyco- protein EnvA. To further investigate the interaction between the TL5 proteins and EnvA, the binding properties of these proteins to EnvA were examined by an ELISA-based binding test. This assay was modified from a previous protocol (23) which allows quick assessment of a large number of Tva mu- tants for their ability to bind EnvA. As shown in Fig. 5, among the 14 TL5 proteins only TL5 chimera 2 displayed a relatively high level of EnvA binding, giving approximately one-third of that of wt Tva, while the remaining 13 TL5 proteins displayed little or no detectable EnvA binding (less than 10% of that of wt Tva). These results suggest that the failure of these TL5 constructs to mediate efficient ASLV-A infection is due to a defect in high-affinity binding to EnvA. It is interesting that although three TL5 constructs (TL5LHW, TL5LHWG, and TL5 chimera 1) supported a low level of ASLV-A infection (Fig. 3 and 4), they did not display detectable EnvA binding (Fig. 5). Rong et al. have previously observed similar discrep- ancies (measured by ELISA-based test) between the ability of many Tva mutants and TL4 proteins to mediate viral entry and FIG. 4. (A) Establishment (using a luciferase gene as the reporter) of the EnvA-pseudotyped HIV system. Human 293T cells were transiently transfected with Tva or mock transfected, and the transfected cells were challenged with the EnvA-pseudotyped HIV viruses. The infected cells were lysed, and luciferase activity levels were measured and are expressed as RLU. EnvA, the ASLV-A glycoprotein-pseudotyped HIV; vector, HIV without any viral glycoprotein. Experiments (performed in triplicate) were repeated several times. The results of a representative experiment are shown here. (B) The abilities of TL5 constructs to mediate ASLV-A entry as measured using the EnvA-pseudotyped HIV viruses. Mock, mock transfected; TL5 Ch2, TL5 chimera 2; TL5 Ch1, TL5 chimera 1. Experiments (performed in triplicate) were repeated three times with consistent results. Bars, standard deviations. VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 687
  • 6. their ability to bind EnvA (22, 23). A plausible explanation for these discrepancies is that the ELISA-based binding test used in this and previous studies is not sensitive enough to distin- guish the binding differences among the TL5 proteins that have low-level binding affinities to EnvA. The spacing between the second and third cysteines of the Tva LDL-A module is important for receptor function. Previ- ously we have shown that the overall conformation of the Tva LDL-A module is different from the structures of the other reported LDL-A modules, including human LDL-A5. The N terminus of the Tva LDL-A module does not contain the signature antiparallel ␤-sheet observed in other LDL-A mod- ules and is more flexible than other reported LDL-A modules (28). Sequence alignment shows that there are nine residues between the second and third cysteines for the Tva LDL-A module and that the other LDL-A modules with reported structures contain only four to six residues. The fact that the TL5 chimera 2 can mediate ASLV-A infection efficiently and TL5LHWG cannot do so prompted us to examine whether the overall conformation of the N terminus of the Tva LDL-A module contributes to the receptor function of Tva. As previ- ously reported, LDL-A22 is the only other known LDL-A module that (just like that of Tva) contains nine residues be- tween C2 and C3, thus conserving the spacing between cys- teines 2 and 3. Wang et al. proposed that LDL-A22 adopts a structure similar to that of the Tva LDL-A module (28). How- ever, none of the nine amino acids in the C2-C3 regions are conserved between human LDL-A22 and the quail Tva LDL-A module, while four of the nine residues are conserved between quail and chicken Tva proteins. Two TL5 variants, C5 and L5, which are identical to TL5 chimera 2 except that the residues between C2 and C3 of TL5 chimera 2 have been replaced with the corresponding residues of chicken Tva (C5) or LDL-A22 of LDL receptor-related protein (L5), respectively, were constructed (Fig. 6A). When they were examined by FACS analysis, both constructs were expressed well on the surface of the transfected 293T cells (data not shown). The EnvA-pseudotyped HIV viruses were used to challenge the transfected 293T cells as described above to examine the TL5 variants for their ability to mediate ASLV-A entry. The cells transiently expressing C5 gave a level of luciferase activity comparable to that of wt Tva and TL5 chimera 2 (Fig. 6B), suggesting that C5 is as efficient as wt Tva and TL5 chimera 2 in mediation of ASLV-A infection. Sur- prisingly, the cells transiently expressing L5 gave a level of luciferase approximately 100-fold higher than the cells express- ing TL5LHWG and only about 6-fold lower than the Tva- expressing cells (Fig. 6B). Since the amino acid sequences of TL5LHWG and L5 are identical between C3 and C6 and are very similar between C1 and C2, we can conclude that the amino acids between C2 and C3 of L5 play an important role in mediation of ASLV-A entry. Furthermore, since only the spacing between C2 and C3 of human LDL-A22 is conserved with respect to that of Tva from quail and chicken, we can conclude that the proper spacing (and thus, likely the proper conformation) of this region plays an important role for the viral receptor function. DISCUSSION In this report we have shown that the putative viral interac- tion residues of Tva that were previously identified by a gain- of-function approach and mutational analysis are indeed crit- ical for the conversion of human LDL-A5 into a functional ASLV-A receptor. Furthermore, our results have revealed a critical role of the N terminus between cysteines 2 and 3 of the FIG. 5. The EnvA-binding abilities of TL5 constructs measured by an ELISA-based binding test. Human 293T cells were transiently transfected with gD-EnvA, myc-Tva, and the TL5 chimeric constructs. An ELISA-based binding test (described in Materials and Methods) was performed to measure the relative level of affinity of binding to EnvA of each of the TL5 proteins compared to that of myc-Tva. Mock, mock transfected. 688 RAI ET AL. J. VIROL.
  • 7. Tva LDL-A module in mediation of ASLV-A entry. Signifi- cantly, our results indicate that the spacing between C2 and C3 of Tva (and thus, the overall conformation of this region) is an important determinant for viral entry. The role of Tva in ASLV-A entry has been extensively ex- amined by molecular, biochemical, and structural analysis. However, most work has concentrated on the C-terminal re- gion of the Tva LDL-A module (C3-C6); this region was found to be essential for viral entry. This has led to the identification of individual residues in this region which are either ligand interaction residues or involved in stability and conformation of the protein (2, 22, 23, 31, 32). For example, Leu34, His38, Trp48, and Gly49 of Tva were identified as being important for viral receptor function. These results are substantiated by the results of the present study. We have shown that replacements of the corresponding residues in human LDL-A5 by these four amino acids, in addition to the amino terminus of Tva, were required to convert human LDL-A5 into a functional viral receptor. Interestingly, the side chains of His38 and Trp48 of Tva were shown to be exposed on the surface, in consistency with the notion that these residues are EnvA contact residues. However, the side chain of Leu34 is buried in the interior of the module together with Phe16 and Pro21, forming the hy- drophobic core. These data indicate that the role of Leu34 is to maintain the correct folding of Tva; thus, it is unlikely to be a ligand contact residue (28). We speculate that conversion of the corresponding residue of human LDL-A5 (Arg19) to a leucine (as described in this study) or conversion of the corre- sponding residue of human LDL-A4 (Ala19) to a leucine (as described in the previous report) can help maintain the correct folding in TL5 and TL4 chimeras, respectively. The major difference between the present study using human LDL-A5 and the previous study using LDL-A4 is that LDL-A5 with the aforementioned four substitutions was not able to mediate efficient viral infection unless its N terminus was also replaced with the corresponding region from Tva. This finding is in contrast to that of the previous study with LDL-A4, which demonstrated that LDL-A4 could be converted into a functional viral receptor with merely the aforementioned substitutions and without re- placement of the N terminus by that of Tva (22). This may be explained by a higher level of sequence homology be- tween Tva and LDL-A4 than between Tva and LDL-A5 at the C terminus. For example, there are two lysine residues in LDL-A5 (K27 and K29) (Fig. 1A) that are absent in the Tva module and LDL-A4. Perhaps these positively charged residues adversely affect the binding affinity between LDL-A5 and EnvA, thus making it a less efficient viral receptor. It should be pointed out that the modified human LDL-A4 protein (TL4G-A19LD23H) was also somewhat defective in EnvA binding compared to that of wt Tva (22). This defect in EnvA binding could be corrected by replacing the N terminus of LDL-A4 with the corresponding region of Tva (data not shown), again suggesting the important role of the N-terminal region of Tva in mediation of efficient EnvA binding and ASLV-A infection. Sequence alignment between Tva and other LDL-A mod- ules indicates that the N terminus of the Tva LDL-A module (between cysteines 1 and 3) is more divergent than the C terminus of other LDL-A modules (Fig. 1A). Indeed, the structure of the LDL-A module of Tva reveals that compared to the other LDL-A module structures reported to date, the N terminus of the Tva module adopts a unique conformation. The Tva LDL-A module does not have the signature anti- parallel ␤-sheet at the N terminus observed in other LDL-A modules; thus, it has an even less recognizable secondary struc- ture (28). Comparisons of quail and chicken Tva protein se- quences indicate that the C1-C2 regions are highly similar except for two conserved variations and that the C3-C6 regions are identical or conserved except for one residue. However, there is significant sequence divergence in the Tva LDL-A FIG. 6. (A) Constructs of TL5 chimera 2 with substituted C2-C3 regions. C5 has the C2-C3 domain of the chicken Tva LDL-A module, and L5 has the C2-C3 domain of the LDL-A22 from LRP2. (B) The abilities of C5 and L5 to mediate ASLV-A infection (as measured using EnvA-pseudotyped HIV infection). Experiments were repeated three times with consistent results. Bars, standard deviations. Mock, mock transfected. VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 689
  • 8. module between quail and chicken in the region between the second and the third cysteines—only four of the nine residues are conserved (Fig. 7). This sequence divergence may dictate subtle differences in this region and determine specificity in receptor usage. Indeed, it was reported that several ASLV-A variants which were genetically selected for a soluble form of quail Tva could not use quail Tva but could still efficiently use chicken Tva as the receptor in viral entry (14–16). These re- sults seem to suggest that specific residues in the N terminus of Tva play an important role in specificity in receptor usage. In contrast, our results have shown that the spacing between the second and third cysteines also plays a critical role in receptor function. The C2-C3 regions of both chicken Tva and LDL-A22 have the same number of residues as the quail Tva module. However, the individual amino acids are more con- served between quail and chicken Tva than between quail Tva and LDL-A22. As mentioned above, four out of nine amino acids between C2 and C3 are conserved between chicken and quail Tva proteins, while not a single residue is conserved between LDL-A22 and quail Tva. Nevertheless, both C5 and L5 were able to mediate viral entry at a level that was at least 100-fold higher than that seen with TL5LHWG (Fig. 6B). Since the C2-C3 regions of both LDL-A4 and LDL-A5 have only four residues (Fig. 7), these results illustrate the impor- tance of maintaining the proper spacing between C2 and C3 of Tva for optimal receptor function. These results may appear to be inconsistent with those of a previous report by Rong et al. demonstrating that deletion of the C1-C3 region of Tva re- duced the viral receptor function of Tva by only approximately 50 to 60%. One possible explanation is that the defective phenotype of the C1-C3 deletion mutant of Tva was masked due to overexpression of the protein in the previous study (23). Another possibility is that there are other viral interaction residues in the C3-C6 region of Tva which are critical for the viral receptor function but which are not conserved in human LDL-A5. Thus, the defective phenotype of TL5 LHWG be- came more apparent than that of the C1-C3 deletion mutant of Tva in mediation of ASLV-A infection. In conclusion, the results of the present study have demon- strated that the spacing between cysteines 2 and 3 of the Tva LDL-A module plays a crucial role in mediation of ASLV-A entry. We hypothesize that this region can help maintain the proper conformation of Tva upon EnvA binding, thus optimiz- ing Tva-EnvA interactions in ASLV-A entry. It is important to point out that the results reported here do not exclude the possibility that some individual residues in this region play a role in viral entry. We have shown that C5 is approximately 10-fold more efficient than L5 in mediation of viral entry (Fig. 6B), suggesting that certain residues of chicken and quail Tva in this region are directly involved in ligand contact. Therefore, the N-terminal region of the Tva LDL-A module is responsible for both correct protein conformation and ligand recognition for optimal receptor function in mediations of ASLV-A entry. ACKNOWLEDGMENTS We thank Judith White, University of Virginia, for providing the polyclonal anti-EnvA tail antibody and Gary Cohen and Roselyn Eisenberg, University of Pennsylvania, for providing the monoclonal anti-gD antibody. We thank Osvaldo Martinez, University of Illinois at Chicago, for help in flow cytometry and Osvaldo Martinez and Balaji Manicassamy for critical reading of the manuscript. The following reagent was obtained through the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID NIH: pNL4-3.Luc.R-E (from Nathaniel Landau). The research work was supported by American Heart Association Midwest Affiliate grant-in-aid 9951134Z and National Institutes of Health grant CA 092459. L.R. was a recipient of the Schweppe Foun- dation Career Development Award. K.R. was supported by a predoc- toral fellowship from American Heart Association Midwest Affiliate. REFERENCES 1. Bates, P., J. A. T. Young, and H. E. Varmus. 1993. A receptor for subgroup A Rous sarcoma virus is related to the low density lipoprotein receptor. Cell 74:1043–1051. 2. Belanger, C., K. Zingler, and J. A. T. Young. 1995. Importance of cysteines in the LDLR-related domains of the subgroup A avian leukosis and sarcoma virus receptor for viral entry. J. Virol. 69:1019–1024. 3. Connolly, L., K. Zingler, and J. A. T. Young. 1994. A soluble form of a receptor for subgroup A avian leukosis and sarcoma virus (ALSV-A) blocks infection and binds directly to ALSV-A. J. Virol. 68:2760–2764. 4. Connor, R. I., B. K. Chen, S. Choe, and N. R. Landau. 1995. Vpr is required for efficient replication of human immunodeficiency virus type-1 in mono- nuclear phagocytes. Virology 206:935–944. 5. Daly, N. L., J. T. Djordjevic, P. A. Kroon, and R. Smith. 1995. Three- dimensional structure of the second cysteine-rich repeat from the human low-density lipoprotein receptor. Biochemistry 34:14474–14481. 6. Daly, N. L., M. J. Scanlon, J. T. Tjordjevic, P. A. Kroon, and R. Smith. 1995. Three-dimensional structure of a cysteine-rich repeat from the low-density lipoprotein receptor. Proc. Natl. Acad. Sci. USA 92:6334–6338. 7. Damico, R. L., J. Crane, and P. Bates. 1998. Receptor-triggered membrane association of a model retroviral glycoprotein. Proc. Natl. Acad. Sci. USA 95:2580–2585. 8. Dolmer, K., W. Huang, and P. G. Gettins. 2000. NMR solution structure of complement-like repeat CR3 from the low density lipoprotein receptor- related protein. Evidence for specific binding to the receptor binding domain of human ␣(2)-macroglobulin. J. Biol. Chem. 275:3264–3269. 9. Fass, D., S. Blacklow, P. S. Kim, and J. M. Berger. 1997. Molecular basis of familial hypercholesterolaemia from structure of LDL receptor module. Nature 388:691–693. 10. Federspiel, M. J., P. Bates, J. A. Young, H. E. Varmus, and S. H. Hughes. 1994. A system for tissue-specific gene targeting: transgenic mice susceptible to subgroup A avian leukosis virus-based retroviral vectors. Proc. Natl. Acad. Sci. USA 91:11241–11245. 11. Gilbert, J. M., L. D. Hernandez, J. W. Balliet, P. Bates, and J. M. White. 1995. Receptor-induced conformational changes in the subgroup A avian leukosis and sarcoma virus envelope glycoprotein. J. Virol. 69:7410–7415. 12. He, J., S. Choe, R. Walker, P. Di Marzio, D. O. Morgan, and N. R. Landau. 1995. Human immunodeficiency virus type 1 viral protein R (Vpr) arrests cells in the G2 phase of the cell cycle by inhibiting p34cdc2 activity. J. Virol. 69:6705–6711. 13. Hernandez, L. D., R. J. Peters, S. E. Delos, J. A. T. Young, D. A. Agard, and J. M. White. 1997. Activation of a retroviral membrane fusion protein: soluble receptor-induced liposome binding of the ALSV envelope glycopro- tein. J. Cell Biol. 139:1455–1464. 14. Holmen, S. L., and M. J. Federspiel. 2000. Selection of a subgroup A avian leukosis virus [ALV(A)] envelope resistant to soluble ALV(A) surface gly- coprotein. Virology 273:364–373. 15. Holmen, S. L., D. C. Melder, and M. J. Federspiel. 2001. Identification of key residues in subgroup A avian leukosis virus envelope determining receptor binding affinity and infectivity of cells expressing chicken or quail Tva re- ceptor. J. Virol. 75:726–737. FIG. 7. Sequence comparison of the C1-C3 regions among the per- tinent LDL-A modules in this study. Residues are numbered at the top according to the mature Tva. Residues identical to those of quail Tva are boxed. 690 RAI ET AL. J. VIROL.
  • 9. 16. Holmen, S. L., D. W. Salter, W. S. Payne, J. B. Dodgson, S. H. Hughes, and M. J. Federspiel. 1999. Soluble forms of the subgroup A avian leukosis virus [ALV(A)] receptor Tva significantly inhibit ALV(A) infection in vitro and in vivo. J. Virol. 73:10051–10060. 17. Huang, W., K. Dolmer, and P. G. Gettins. 1999. NMR solution structure of complement-like repeat CR8 from the low density lipoprotein receptor- related protein. J. Biol. Chem. 274:14130–14136. 18. Kozarsky, K., M. Grossman, and J. M. Wilson. 1993. Adenovirus-mediated correction of the genetic defect in hepatocytes from patients with familial hypercholesterolemia. Somat. Cell Mol. Genet. 19:449–458. 19. Mothes, W., A. L. Boerger, S. Narayan, J. M. Cunningham, and J. A. T. Young. 2000. Retroviral entry mediated by receptor priming and low pH triggering of an envelope glycoprotein. Cell 103:679–689. 20. North, C. L., and S. C. Blacklow. 2000. Solution structure of the sixth LDL-A module of the LDL receptor. Biochemistry 39:2564–2571. 21. Rong, L., and P. Bates. 1995. Analysis of the subgroup A avian sarcoma and leukosis virus receptor: the 40-residue, cysteine-rich, low-density lipoprotein receptor repeat motif of Tva is sufficient to mediate viral entry. J. Virol. 69:4847–4853. 22. Rong, L., K. Gendron, and P. Bates. 1998. Conversion of a human low- density lipoprotein receptor ligand-binding repeat to a virus receptor: iden- tification of residues important for ligand specificity. Proc. Natl. Acad. Sci. USA 95:8467–8472. 23. Rong, L., K. Gendron, B. Strohl, R. Shenoy, R. J. Wool-Lewis, and P. Bates. 1998. Characterization of determinants for envelope binding and infection in Tva, the subgroup A avian sarcoma and leukosis virus receptor. J. Virol. 72:4552–4559. 24. Rudenko, G., L. Henry, K. Henderson, K. Ichtchenko, M. S. Brown, J. L. Goldstein, and J. Deisenhofer. 2002. Structure of the LDL receptor extra- cellular domain at endosomal pH. Science 298:2353–2358. 25. Simonovic, M., K. Dolmer, W. Huang, D. K. Strickland, K. Volz, and P. G. Gettins. 2001. Calcium coordination and pH dependence of the calcium affinity of ligand-binding repeat CR7 from the LRP. Comparison with re- lated domains from the LRP and the LDL receptor. Biochemistry 40:15127– 15134. 26. Tonelli, M., R. J. Peters, T. L. James, and D. A. Agard. 2001. The solution structure of the viral binding domain of Tva, the cellular receptor for sub- group A avian leukosis and sarcoma virus. FEBS Lett. 509:161–168. 27. Wang, Q. Y., K. Dolmer, W. Huang, P. G. Gettins, and L. Rong. 2001. Role of calcium in protein folding and function of Tva, the receptor of subgroup A avian sarcoma and leukosis virus. J. Virol. 75:2051–2058. 28. Wang, Q. Y., W. Huang, K. Dolmer, P. G. Gettins, and L. Rong. 2002. Solution structure of the viral receptor domain of Tva and its implications in viral entry. J. Virol. 76:2848–2856. 29. Young, J. A. T., P. Bates, and H. E. Varmus. 1993. Isolation of a chicken gene that confers susceptibility to infection by subgroup A avian leukosis and sarcoma viruses. J. Virol. 67:1811–1816. 30. Yu, X., Q. Y. Wang, Y. Guo, K. Dolmer, J. A. Young, P. G. Gettins, and L. Rong. 2003. Kinetic analysis of binding interaction between the subgroup A Rous sarcoma virus glycoprotein SU and its cognate receptor Tva: calcium is not required for ligand binding. J. Virol. 77:7517–7526. 31. Zingler, K., and J. A. T. Young. 1996. Residue Trp-48 of Tva is critical for viral entry but not for high-affinity binding to the SU glycoprotein of sub- group A avian leukosis and sarcoma viruses. J. Virol. 70:7510–7516. 32. Zingler, K., C. Belanger, R. Peters, D. Agard, and J. A. T. Young. 1995. Identification and characterization of the viral interaction determinants of the subgroup A avian leukosis virus receptor. J. Virol. 69:4261–4266. VOL. 78, 2004 ROLE OF Tva IN ASLV-A ENTRY 691