ArticlePDF Available

The TRIM5 Genotype of Rhesus Macaques Affects Acquisition of Simian Immunodeficiency Virus SIVsmE660 Infection after Repeated Limiting-Dose Intrarectal Challenge

American Society for Microbiology
Journal of Virology
Authors:

Abstract and Figures

It has recently been shown that polymorphism at the rhesus macaque TRIM5 locus can affect simian immunodeficiency virus (SIV) replication. Here we show that TRIM5 alleles can also affect acquisition of SIVsmE660. Animals coexpressing the TRIM5TFP and TRIM5CypA alleles took significantly longer to become infected with SIVsmE660, but not SIVmac239, after repeated limiting-dose intrarectal challenge than did animals expressing other TRIM5 allele combinations. Our results indicate that the TRIM5 alleles can be a barrier to productive infection and that this should be taken into account when designing acquisition studies using SIVsmE660 or related viruses.
Content may be subject to copyright.
JOURNAL OF VIROLOGY, Sept. 2011, p. 9637–9640 Vol. 85, No. 18
0022-538X/11/$12.00 doi:10.1128/JVI.05074-11
Copyright © 2011, American Society for Microbiology. All Rights Reserved.
NOTES
The TRIM5 Genotype of Rhesus Macaques Affects Acquisition of
Simian Immunodeficiency Virus SIVsmE660 Infection after
Repeated Limiting-Dose Intrarectal Challenge
Matthew R. Reynolds,
1
* Jonah B. Sacha,
2,3
Andrea M. Weiler,
1
Gretta J. Borchardt,
1
Chrystal E. Glidden,
1
Neil C. Sheppard,
4
Francesca A. Norante,
1
Philip A. Castrovinci,
1
Jacqueline J. Harris,
5
Henry T. Robertson,
5
Thomas C. Friedrich,
1
Adrian B. McDermott,
6
Nancy A. Wilson,
1
David B. Allison,
5
Wayne C. Koff,
6
Welkin E. Johnson,
7
and David I. Watkins
1,8
AIDS Vaccine Research Laboratory, 555 Science Dr., Madison, Wisconsin 53711
1
; Vaccine and Gene Therapy Institute, Oregon Health and
Science University, 505 NW 185th Ave., Beaverton, Oregon 97006
2
; Oregon National Primate Research Center, Oregon Health and
Science University, 505 NW 185th Ave., Beaverton, Oregon 97006
3
; Vaccine Research, Worldwide Research and Development,
Pfizer Inc., 10777 Science Center Drive, San Diego, California 92121
4
; Department of Biostatistics, Section on Statistical Genetics,
University of Alabama at Birmingham, Birmingham, Alabama 35294
5
; International AIDS Vaccine Initiative, New York,
New York 10038
6
; New England Primate Research Center, Department of Microbiology and Molecular Genetics,
Harvard Medical School, Southborough, Massachusetts
7
; and Department of Pathology and
Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin
8
Received 10 May 2011/Accepted 27 June 2011
It has recently been shown that polymorphism at the rhesus macaque TRIM5 locus can affect simian
immunodeficiency virus (SIV) replication. Here we show that TRIM5 alleles can also affect acquisition of
SIVsmE660. Animals coexpressing the TRIM5
TFP
and TRIM5
CypA
alleles took significantly longer to become
infected with SIVsmE660, but not SIVmac239, after repeated limiting-dose intrarectal challenge than did
animals expressing other TRIM5 allele combinations. Our results indicate that the TRIM5 alleles can be a
barrier to productive infection and that this should be taken into account when designing acquisition studies
using SIVsmE660 or related viruses.
Lentiviruses and their hosts have been interacting and
adapting to each other for millions of years. Humans and
nonhuman primates, like other mammals, express cellular re-
striction factors to inhibit viral replication (6). These restric-
tion factors offer protection against a broad range of viruses
and represent an important barrier to zoonotic transmissions.
Recently, three major classes of retroviral restriction factors
have been identified: APOBEC3, tripartite motif protein 5
alpha (TRIM5), and tetherin (12, 16, 19, 20). Each of these
factors targets a different step in the virus life cycle to suppress
viral replication. To counteract these restriction factors, pri-
mate lentiviruses have developed a series of accessory proteins
that, in part, circumvent their actions and allow viruses to
efficiently replicate (11).
Polymorphic alleles at the rhesus macaque TRIM5 locus cor-
relate with a 1.3- to 3-log reduction in simian immunodeficiency
virus (SIV) replication (7, 10). The mechanism by which TRIM5
affects viral replication is unclear, but it appears to act by binding
incoming viral capsids and mediating premature viral uncoating
(18). A recent study also suggests a role for TRIM5 mediating the
early, innate immune response to retroviral infection (14). Rhesus
macaque TRIM5 is polymorphic, and the encoded protein in-
cludes a TRIM5-cyclophilin A (CypA) chimera in which CypA
replaced the TRIM5 binding domain (7, 10, 13, 17, 19). While
there are multiple TRIM5 alleles, they can be categorized into
three functional allelic classes based on variations in their C-ter-
minal domains and their relative effects on SIVsmE543-3 repli-
cation: TRIM5
CypA
, TRIM5
TFP
, and TRIM5
Q
(7). Combinations
of these alleles result in six possible functional TRIM5 genotypes.
A previous study showed that cell lines expressing TRIM5
TFP
or
TRIM5
CypA
alleles were resistant to infection with the molecular
clone SIVsmE543-3 but not with SIVmac239 (7). Cells expressing
TRIM5
Q
alleles were susceptible to both viruses. These in vitro
results were recapitulated in vivo in a cohort of animals infected
with SIVsmE543-3. Animals with two resistance-conferring
TRIM5 alleles (TRIM5
TFP/TFP
or TRIM5
TFP/CypA
) had a 2.0- to
3.0-log reduction in chronic-phase plasma virus replication in
comparison to animals with the TRIM5
Q/Q
genotype. Further-
more, animals with one resistance-conferring allele (TRIM5
TFP/Q
or TRIM5
Q/CypA
) had intermediate plasma viral loads. A less
dramatic effect on plasma virus replication was also seen in a
cohort of SIVmac251-infected animals by using similar TRIM5
groupings (10).
We first investigated whether the biological isolate
SIVsmE660 might harbor TRIM5-resistant variants. Previous
* Corresponding author. Mailing address: AIDS Vaccine Research
Laboratory, 555 Science Dr., Madison, WI 53711. Phone: (608) 890-
0842. Fax: (608) 265-8084. E-mail: mrreynol@wisc.edu.
Published ahead of print on 6 July 2011.
9637
studies identified amino acids in Gag p27
CA
affecting TRIM5-
mediated restriction (2, 7, 9). Since two different stocks of
SIVsmE660 were used in our studies, we employed ultradeep
pyrosequencing as previously described (1) to determine
whether there existed any variation that might result in
TRIM5-resistant quasispecies. Briefly, we used four overlap-
ping reverse transcription-PCR amplicons of approximately 2.5
kb spanning the entire SIVsmE660 genome. The reverse tran-
scription-PCR products were then randomly fragmented using
modified transposons (Nextera; Epicentre Biotechnology) and
pyrosequenced. At average nucleotide depths of 493 for
SIVsmE660 stock 17480 and 819 for SIVsmE660 stock 17571,
we found no nonsynonymous variation above 1% from the
SIVsmE543-3 regions previously identified to affect rhesus ma-
caque TRIM5-mediated restriction (Fig. 1) (7). This suggested
that an animal’s TRIM5 genotype could impose a bottleneck
for productive infection following limiting-dose intrarectal
SIVsmE660 challenge.
Only a few human immunodeficiency virus (HIV) strains
cross mucosal barriers to establish infection in humans (4).
Investigators have been using repeated limiting-dose SIV chal-
lenge of rhesus macaques to mimic HIV transmission and have
shown that only one to three viral species similarly initiate
infection in monkeys (5, 21). Under these conditions, it is
possible that the TRIM5 genotype of an animal might play a
role in whether an animal becomes productively infected with
SIV. To address this issue, we retrospectively analyzed a cohort
of 62 Indian rhesus macaques at the Wisconsin National Pri-
mate Research Center (WNPRC) that were not vaccinated
with SIV antigens to determine if TRIM5 genotype affected
the rate at which they became productively infected with
SIVsmE660. We genotyped our animals based on the following
three allelic classes: TRIM5
CypA
, TRIM5
TFP
, and TRIM5
Q
.We
established the TRIM5 genotypes of the animals either by
sequencing genomic DNA as previously described (7) or by
sequence-specific priming PCR (PCR-SSP) assays. To distin-
guish TRIM5
TFP
and TRIM5
Q
alleles by PCR-SSP, we took
advantage of unique polymorphisms in the TRIM5 SPRY/
B30.2 domain and used previously published thermal cycling
conditions and reagents (3). The TRIM5
TFP
and TRIM5
Q
PCR-SSP reactions share a forward primer (5-AGA GAG
CTA ACA GAT GCC CG-3) designed to anneal to both of
the TRIM5 variants, while the reverse primers targeted unique
sites within the TRIM5
TFP
and TRIM5
Q
alleles to confer spec
-
ificity (TFP allele reverse primer, 5-ACA ATG AAA GGA
GCA AAA GGA GTA TGT G-3; Q allele reverse primer,
5-CAC AAT GAA AGG AGC AAA AGG AGT ATG TA-
3). Primers were used at a 0.3 M working concentration. The
TRIM5
CypA
allele was genotyped using a previously published
protocol which utilized PCR followed by NsiI digestion (13).
To confirm that the consistency of the PCR-SSP genotyping
was equivalent to that of genomic DNA sequencing, we tested
a subset of animals by both methods and obtained identical
results.
We next investigated whether the TRIM5 genotype affected
acquisition of SIVsmE660. Rhesus macaques used in this anal-
ysis were intrarectally inoculated on a weekly or biweekly basis
with initial limiting doses ranging from 225 to 450 50% tissue
culture infective doses (TCID
50
) for SIVsmE660-17571 (52
animals) and 800 TCID
50
for SIVsmE660-17480 (10 animals)
as previously described (15, 21). We performed a Kaplan-
Meier analysis to test whether any of the TRIM5 genotypes
differed in the number of challenges required to achieve pro-
ductive infection. We discovered that animals having the
TRIM5
TFP/CypA
genotype took significantly more exposures to
get infected than animals expressing the other TRIM5 geno-
types (log rank test; chi-square 15.74; df 5; P 0.008)
(analyses performed with GraphPad Prism version 4.0c) (Fig.
2A). Of note, four animals with the TRIM5
TFP/CypA
genotype
were the only animals to remain uninfected at the conclusion
of intrarectal challenges, two animals after the seventh chal-
lenge and two after the eleventh challenge. We therefore fur-
ther explored the resistance to infection with SIVsmE660 of
the TRIM5
TFP/CypA
animals in comparison to the other geno
-
types. We compared the numbers of challenges needed to
achieve productive infection per TRIM5 genotype by using the
parametric generalized gamma model because it was deter-
mined to be the best model in comparison to nonparametric
models by Akaike information criterion scores for examining
dosages that increased over time. For this analysis, we desig-
nated the inoculation after intrarectal challenges were stopped
for the uninfected animals as the infecting dose (e.g., we des-
ignated the animals remaining uninfected after 11 inoculations
as being infected with the 12th inoculation). Animals with the
TRIM5
TFP/CypA
genotype took significantly longer to produc
-
tively infect with limiting doses of SIVsmE660 than each of the
remaining TRIM5 genotypes (P 0.0001) (Fig. 2B).
Since the TRIM5 genotype affects acquisition of infection
with SIVsmE660, we sought to determine if it has the same
effect on another SIV strain commonly used in challenge stud-
ies, the molecularly cloned SIVmac239. Kirmaier et al. previ-
ously established that SIVmac239, unlike SIVsmE543-3, is re-
sistant to all of the TRIM5 allelic classes in vitro, which is likely
due to amino acid substitutions in the viral capsid that prevent
or inhibit TRIM5-mediated restriction (7). However, it is still
possible in the setting of a limiting-dose challenge in which
only a few viruses likely cross the mucosal barrier and establish
infection that TRIM5 might have an effect on acquisition. We
therefore examined a cohort of 38 naïve animals repeatedly
challenged with limiting doses of SIVmac239 (produced in
vitro with rhesus macaque peripheral blood mononuclear cells
[PBMC]) housed at the WNPRC and at the University of
FIG. 1. Sequence of SIVsmE660 Gag p27
CA
known to affect TRIM5-
mediated restriction. Partial sequences of the core nucleocapsid protein
amino acids 72 to 101 of the two stocks of SIVsmE660 analyzed in this
study are aligned against that of the closely related molecular clone
SIVsmE543-3. The putative amino acids affecting restriction mediated by
TRIM5
TFP
(
74
LPA
76
) and TRIM5
CypA
(R
83
) are highlighted in red. Non
-
synonymous variations from the SIVsmE543-3 sequence are color coded
by frequency. X indicates a mixed amino acid sequence.
9638 NOTES J. V
IROL.
Pittsburgh. Of note, a few animals included in this cohort
were repeatedly challenged with either 30 or 300 TCID
50
SIVmac239, which in retrospect was probably an extremely
limiting dose of the virus, because it required between 8 and 21
challenges to achieve infection. We also had two animals with
either the TRIM5
TFP/TFP
or TRIM5
TFP/Q
genotype that did not
become infected after 13 limiting-dose challenges. Despite
this, we detected no significant difference in the rates of ac-
quisition of SIVmac239 for the different TRIM5 genotypes
when we performed the Kaplan-Meier analysis (P 0.67) (Fig.
3). However, due to the relatively small number of animals in
this analysis, it is possible that TRIM5 has a more limited effect
on acquisition of SIVmac239 that cannot be discerned in our
study.
Our study highlights the complex nature of virus-host inter-
actions and the difficulty of modeling HIV transmission in
rhesus macaques. Our analysis also expands upon a recent
report showing that TRIM5 alleles can affect the acquisition
of SIVsmE660 infection by naïve and vaccinated rhesus ma-
caques (8). In this study, however, TRIM5
TFP
and TRIM5
CypA
were grouped together as “restrictive” alleles, and homologous
expression of these alleles resulted in delayed infection with
SIVsmE660. In our cohort of animals, we further subdivided
the genotypes to include TRIM5
CypA
as a separate allele and
only the heterozygous TRIM5
TFP/CypA
genotype made a signif
-
icant impact on the rate of acquisition of SIVsmE660 infection.
It is possible that since animals with the TRIM5
TFP/CypA
geno
-
type have two TRIM5 molecules that bind distinct regions of
FIG. 2. Acquisition of SIVsmE660 infection after repeated limiting-dose challenge is affected by TRIM5 genotype. (A) Kaplan-Meier curve
analysis of the effects different TRIM5 genotypes have on the rate of acquisition of SIVsmE660 infection after repeated limiting-dose intrarectal
challenge. The statistical significance of the rates of infection of the different TRIM5 genotypes was determined by log rank test. (B) A comparison
of the numbers of challenges needed to productively infect animals expressing the different TRIM5 genotypes with SIVsmE660. Significance
between the rates of infection of animals having the TRIM5
TFP/CypA
genotype and the other TRIM5 genotypes was determined by parametric
generalized gamma model.
FIG. 3. Acquisition of SIVmac239 infection after repeated limiting-dose intrarectal challenge is not affected by TRIM5 genotype. The
Kaplan-Meier curve analysis revealed no significant differences in the acquisition of SIVmac239 infection after repeated limiting-dose intrarectal
challenge among the TRIM5 genotypes. We performed a power analysis and determined that our cohort of SIVmac239 challenged animals had
73% power to detect an effect by the TRIM5
TFP/CypA
genotype on acquisition of infection similar to that observed for the cohort of SIVsmE660
challenged animals.
V
OL. 85, 2011 NOTES 9639
the viral capsid, they eliminate incoming virus more effectively
than animals having TRIM5 molecules targeting only one
region of the viral capsid, even for animals homozygous for
resistance-conferring TRIM5 alleles. It is also possible that the
TRIM5
TFP/TFP
and TRIM5
CypA/CypA
genotypes have a less dra
-
matic effect on the acquisition of infection, which would be-
come apparent in a larger cohort of animals. Our study also
indicates that under physiological conditions in which only a
few viruses are crossing the mucosal barrier, TRIM5 can
decrease the probability of productive infection occurring.
However, since restriction by TRIM5 is known to be satura-
ble, this effect may be subtle and overcome by high-dose mu-
cosal or intravenous exposures (19). In this regard, the previ-
ous studies identifying that TRIM5 genotypes differentially
affects SIVsmE543-3 and SIVmac251 replication did not note
an effect on acquisition of infection after intravenous inocula-
tion (7, 10). Our results suggest that animals entering repeated
limiting-dose SIV challenge studies should be typed for their
TRIM5 genotype, especially if SIVsmE660 or a related virus is
used. When possible, TRIM5
TFP/CypA
animals should be either
excluded from the study or balanced among test groups in a
manner similar to that for animals expressing protective major
histocompatibility complex (MHC) class I alleles.
We thank the veterinary staff at the Wisconsin National Primate
Research Center (WNPRC) for their assistance. We also thank Ben
Burwitz, Ben Bimber, Simon Lank, and David O’Connor for their
guidance in pyrosequencing and analyzing the SIVsmE660 stocks.
This research was supported by funds from the International AIDS
Vaccine Initiative, National Institutes of Health (NIH) grants R01
AI049120, R01 AI076114, R01 AI076114, R24 RR016038, R24
RR015371, and R37 AI076114 and contract HHSN266200400088C to
D.I.W., grant R01 AI083118 to W.E.J., grant P51 RR000167 to the
WNPRC, and grant RR00168 to the New England Primate Research
Center (NEPRC) from the National Center for Research Resources
(NCRR), a component of the NIH. This research was also supported
by Pfizer Inc.-sponsored research agreement 140-N-2076312 with
J.B.S. Additionally, this research was conducted at a facility con-
structed with support from Research Facilities Improvement Program
grant numbers RR15459-01 and RR020141-01.
This publication’s contents are solely the responsibility of the au-
thors and do not necessarily represent the official views of NCRR or
NIH.
We have no conflicting financial interests.
REFERENCES
1. Bimber, B. N., et al. 2010. Whole-genome characterization of human and
simian immunodeficiency virus intrahost diversity by ultradeep pyrosequenc-
ing. J. Virol. 84:12087–12092.
2. Hatziioannou, T., S. Cowan, U. K. Von Schwedler, W. I. Sundquist, and P. D.
Bieniasz. 2004. Species-specific tropism determinants in the human immu-
nodeficiency virus type 1 capsid. J. Virol. 78:6005–6012.
3. Kaizu, M., et al. 2007. Molecular typing of major histocompatibility complex
class I alleles in the Indian rhesus macaque which restrict SIV CD8 T cell
epitopes. Immunogenetics 59:693–703.
4. Keele, B. F., et al. 2008. Identification and characterization of transmitted
and early founder virus envelopes in primary HIV-1 infection. Proc. Natl.
Acad. Sci. U. S. A. 105:7552–7557.
5. Keele, B. F., et al. 2009. Low-dose rectal inoculation of rhesus macaques by
SIVsmE660 or SIVmac251 recapitulates human mucosal infection by HIV-1.
J. Exp. Med. 206:1117–1134.
6. Kirchhoff, F. 2010. Immune evasion and counteraction of restriction factors
by HIV-1 and other primate lentiviruses. Cell Host Microbe 8:55–67.
7. Kirmaier, A., et al. 2010. TRIM5 suppresses cross-species transmission of a
primate immunodeficiency virus and selects for emergence of resistant vari-
ants in the new species. PLoS Biol. 8:pii:e1000462.
8. Letvin, N. L., et al. 2011. Immune and genetic correlates of vaccine protec-
tion against mucosal infection by SIV in monkeys. Sci. Transl. Med. 3:81ra36.
9. Li, Y., X. Li, M. Stremlau, M. Lee, and J. Sodroski. 2006. Removal of
arginine 332 allows human TRIM5alpha to bind human immunodeficiency
virus capsids and to restrict infection. J. Virol. 80:6738–6744.
10. Lim, S. Y., et al. 2010. TRIM5alpha modulates immunodeficiency virus
control in rhesus monkeys. PLoS Pathog. 6:e1000738.
11. Malim, M. H., and M. Emerman. 2008. HIV-1 accessory proteins–ensuring
viral survival in a hostile environment. Cell Host Microbe 3:388–398.
12. Neil, S. J., T. Zang, and P. D. Bieniasz. 2008. Tetherin inhibits retrovirus
release and is antagonized by HIV-1 Vpu. Nature 451:425–430.
13. Newman, R. M., et al. 2008. Evolution of a TRIM5-CypA splice isoform in
old world monkeys. PLoS Pathog. 4:e1000003.
14. Pertel, T., et al. 2011. TRIM5 is an innate immune sensor for the retrovirus
capsid lattice. Nature 472:361–365.
15. Reynolds, M. R., et al. 2010. Macaques vaccinated with simian immunode-
ficiency virus SIVmac239Delta nef delay acquisition and control replication
after repeated low-dose heterologous SIV challenge. J. Virol. 84:9190–9199.
16. Sheehy, A. M., N. C. Gaddis, J. D. Choi, and M. H. Malim. 2002. Isolation
of a human gene that inhibits HIV-1 infection and is suppressed by the viral
Vif protein. Nature 418:646–650.
17. Song, B., et al. 2005. The B30.2(SPRY) domain of the retroviral restriction
factor TRIM5alpha exhibits lineage-specific length and sequence variation in
primates. J. Virol. 79:6111–6121.
18. Strebel, K., J. Luban, and K. T. Jeang. 2009. Human cellular restriction
factors that target HIV-1 replication. BMC Med. 7:48.
19. Stremlau, M., et al. 2004. The cytoplasmic body component TRIM5alpha
restricts HIV-1 infection in Old World monkeys. Nature 427:848–853.
20. Van Damme, N., et al. 2008. The interferon-induced protein BST-2 restricts
HIV-1 release and is downregulated from the cell surface by the viral Vpu
protein. Cell Host Microbe 3:245–252.
21. Wilson, N. A., et al. 2009. Vaccine-induced cellular responses control simian
immunodeficiency virus replication after heterologous challenge. J. Virol.
83:6508–6521.
9640 NOTES J. VIROL.
... Such diversity at the TRIM5 locus is of interest given the frequency with which different NHP species and subspecies have been used in HIV research, with TRIM5α polymorphism demonstrated to influence SIV susceptibility in rhesus macaques [108,109]. Moreover, TRIM5 polymorphism represents a key host factor in determining cross-species transmission with impacts on virus pressure exerted by the host driving variant emergence [110]. ...
... Specifically, in experimental SIV challenge studies, acute and steady-state viraemia in plasma represents a major readout of virus infection facilitating the interpretation of the efficacy of vaccine and/or anti-viral treatments. Polymorphisms in TRIM5α were correlated with differential viraemic control of wild-type SIV infection, particularly in Rhesus macaques [59,103,108,109]. Hence, specific TRIM5α alleles may need to be specifically excluded from studies or their effects minimised by balancing among test and control groups. ...
Article
Full-text available
Understanding infectious disease pathogenesis and evaluating novel candidate treatment interventions for human use frequently requires prior or parallel analysis in animal model systems. While rodent species are frequently applied in such studies, there are situations where non-human primate (NHP) species are advantageous or required. These include studies of animals that are anatomically more akin to humans, where there is a need to interrogate the complexity of more advanced biological systems or simply reflect susceptibility to a specific infectious agent. The contribution of different arms of the immune response may be addressed in a variety of NHP species or subspecies in specific physiological compartments. Such studies provide insights into immune repertoires not always possible from human studies. However, genetic variation in outbred NHP models may confound, or significantly impact the outcome of a particular study. Thus, host factors need to be considered when undertaking such studies. Considerable knowledge of the impact of host immunogenetics on infection dynamics was elucidated from HIV/SIV research. NHP models are now important for studies of emerging infections. They have contributed to delineating the pathogenesis of SARS-CoV-2/COVID-19, which identified differences in outcomes attributable to the selected NHP host. Moreover, their use was crucial in evaluating the immunogenicity and efficacy of vaccines against COVID-19 and establishing putative correlates of vaccine protection. More broadly, neglected or highly pathogenic emerging or re-emergent viruses may be studied in selected NHPs. These studies characterise protective immune responses following infection or the administration of candidate immunogens which may be central to the accelerated licensing of new vaccines. Here, we review selected aspects of host immunogenetics, specifically MHC background and TRIM5 polymorphism as exemplars of adaptive and innate immunity, in commonly used Old and New World host species. Understanding this variation within and between NHP species will ensure that this valuable laboratory source is used most effectively to combat established and emerging virus infections and improve human health worldwide.
... The first one is the possibility that host factors other than adaptive immune responses contributed to the group A vaccinees' resistance to SIV infection. Certain combinations of TRIM5 alleles, for example, are associated with intrinsic resistance to mucosal challenge with SIVsmE660 in RMs (41,42). However, because the SIVmac239 capsid protein is refractory to TRIM5a-mediated restriction, expression of SIVsmE660-restrictive TRIM5 alleles has not been linked to delayed SIVmac239 acquisition in rectally challenged RMs (42). ...
... Certain combinations of TRIM5 alleles, for example, are associated with intrinsic resistance to mucosal challenge with SIVsmE660 in RMs (41,42). However, because the SIVmac239 capsid protein is refractory to TRIM5a-mediated restriction, expression of SIVsmE660-restrictive TRIM5 alleles has not been linked to delayed SIVmac239 acquisition in rectally challenged RMs (42). A homozygous 32-bp deletion in the CCR5 chemokine receptor gene (i.e., the CCR5D32 allele) that abrogates cell surface expression of CCR5 is also known to confer potent resistance to R5-tropic HIV infection in humans (43). ...
Article
Full-text available
A prophylactic vaccine that confers durable protection against human immunodeficiency virus (HIV) would provide a valuable tool to prevent new HIV/AIDS cases. As herpesviruses establish lifelong infections that remain largely subclinical, the use of persistent herpesvirus vectors to deliver HIV antigens may facilitate the induction of long-term anti-HIV immunity. We previously developed recombinant (r) forms of the gamma-herpesvirus rhesus monkey rhadinovirus (rRRV) expressing a replication-incompetent, near-full-length simian immunodeficiency virus (SIVnfl) genome. We recently showed that 8/16 rhesus macaques (RMs) vaccinated with a rDNA/rRRV-SIVnfl regimen were significantly protected against intrarectal (IR) challenge with SIVmac239. Here we investigated the longevity of this vaccine-mediated protection. Despite receiving no additional booster immunizations, the protected rDNA/rRRV-SIVnfl vaccinees maintained detectable cellular and humoral anti-SIV immune responses for more than 1.5 years after the rRRV boost. To assess if these responses were still protective, the rDNA/rRRV-SIVnfl vaccinees were subjected to a second round of marginal-dose IR SIVmac239 challenges, with eight SIV-naïve RMs serving as concurrent controls. After three SIV exposures, 8/8 control animals became infected, compared to 3/8 vaccinees. This difference in SIV acquisition was statistically significant ( P = 0.0035). The three vaccinated monkeys that became infected exhibited significantly lower viral loads than those in unvaccinated controls. Collectively, these data illustrate the ability of rDNA/rRRV-SIVnfl vaccination to provide long-term immunity against stringent mucosal challenges with SIVmac239. Future work is needed to identify the critical components of this vaccine-mediated protection and the extent to which it can tolerate sequence mismatches in the challenge virus. IMPORTANCE We report on the long-term follow-up of a group of rhesus macaques (RMs) that received an AIDS vaccine regimen and were subsequently protected against rectal acquisition of simian immunodeficiency virus (SIV) infection. The vaccination regimen employed included a live recombinant herpesvirus vector that establishes persistent infection in RMs. Consistent with the recurrent SIV antigen expression afforded by this herpesvirus vector, vaccinees maintained detectable SIV-specific immune responses for more than 1.5 years after the last vaccination. Importantly, these vaccinated RMs were significantly protected against a second round of rectal SIV exposures performed one year after the first SIV challenge phase. These results are relevant for HIV vaccine development because they show the potential of herpesvirus-based vectors to maintain functional antiretroviral immunity without the need for repeated boosting.
... WES datasets provide investigators with the capability to examine sequence variants across the entire coding region of the genome. Although SIV researchers have traditionally focused on specific MHC genotypes that are associated with exceptional control of viral replication, there is also interest in evaluating variation in host restriction factor genes, such as TRIM5, Tetherin, and APOBEC3G receptor genes (Reynolds et al. 2011;Janaka et al. 2018;Krupp et al. 2013;Ericsen et al. 2014). Exome sequencing will also provide an opportunity for investigators in multiple fields to evaluate variants from a wide variety of pathways that are important for biomedical research (Haus et al. 2014). ...
... Exome-wide datasets offer the promise of retrospectively identifying candidate DNA sequence variants that may be responsible for unexpected experimental outcomes in studies with macaques and other nonhuman primates. The availability of exome-wide sequences in conjunction with MHC genotypes may also increase the rigor of prospective macaque experiments by enabling more sophisticated balancing of experimental groups, and exclusion of animals whose genetics are likely to strongly bias experimental results Reynolds et al. 2011;Haus et al. 2014). ...
Article
Full-text available
Indian rhesus macaque major histocompatibility complex (MHC) variation can influence the outcomes of transplantation and infectious disease studies. Frequently, rhesus macaques are MHC genotyped to identify variants that could account for unexpected results. Since the MHC is only one region in the genome where variation could impact experimental outcomes, strategies for simultaneously profiling variation in the macaque MHC and the remainder of the protein coding genome would be useful. Here we determine MHC class I and class II genotypes using target-capture probes enriched for MHC sequences, a method we term macaque exome sequence (MES) genotyping. For a cohort of 27 Indian rhesus macaques, we describe two methods for obtaining MHC genotypes from MES data and demonstrate that the MHC class I and class II genotyping results obtained with these methods are 98.1% and 98.7% concordant, respectively, with expected MHC genotypes. In contrast, conventional MHC genotyping results obtained by deep sequencing of short multiplex PCR amplicons were only 92.6% concordant with expectations for this cohort.
... The susceptibility of SIVsmE543-3, and SIVsmE660, to certain rhesus macaque TRIM5 proteins, is an essential factor to be considered in using these challenge viruses. Several investigators are now working to adapt SIVsmE543-3 and SIVsmE660 to restrictive rhesus macaque TRIM5 variants to generate challenge viruses with more uniform resistance in this model [314]. ...
Article
Full-text available
Various research studies that have investigated the association between HIV infection and addiction underpin the role of various drugs of abuse in impairing immunological and non-immunological pathways of the host system, ultimately leading to augmentation of HIV infection and disease progression. These studies have included both in vitro and in vivo animal models wherein investigators have assessed the effects of various drugs on several disease parameters to decipher the impact of drugs on both HIV infection and progression of HIV-associated neurocognitive disorders (HAND). However, given the inherent limitations in the existing animal models of HAND, these investigations only recapitulated specific aspects of the disease but not the complex human syndrome. Despite the inability of HIV to infect rodents over the last 30 years, multiple strategies have been employed to develop several rodent models of HAND. While none of these models can accurately mimic the overall pathophysiology of HAND, they serve the purpose of modeling some unique aspects of HAND. This review provides an overview of various animal models used in the field and a careful evaluation of methodological strengths and limitations inherent in both the model systems and study designs to understand better how the various animal models complement one another.
... Similar to the M19 study, animals in the M15 study were subjected to weekly intravaginal SIVsmE660 challenges (12 maximum) to determine vaccine efficacy. As we and others reported previously for intrarectal challenges (20,(22)(23)(24)(25), the protection against intravaginal SIVsmE660 was significantly influenced by the TRIM5 genotype and was mainly observed in TRIM5-restrictive animals but not in permissive animals ( fig.S4, A to C). In addition, the group sizes were too small to be able to determine the influence of vaccineinduced IFN + CD4 T cell response on vaccine efficacy. ...
Article
Full-text available
Activated CD4 T cells are a major target of HIV infection. Results from the STEP HIV vaccine trial highlighted a potential role for total activated CD4 T cells in promoting HIV acquisition. However, the influence of vaccine insert-specific CD4 T cell responses on HIV acquisition is not known. Here, using the data obtained from four macaque studies, we show that the DNA prime/modified vaccinia Ankara boost vaccine induced interferon γ (IFNγ ⁺ ) CD4 T cells [T helper 1 (T H 1) cells] rapidly migrate to multiple tissues including colon, cervix, and vaginal mucosa. These mucosal T H 1 cells persisted at higher frequencies and expressed higher density of CCR5, a viral coreceptor, compared to cells in blood. After intravaginal or intrarectal simian immunodeficiency virus (SIV)/simian-human immunodeficiency virus (SHIV) challenges, strong vaccine protection was evident only in animals that had lower frequencies of vaccine-specific T H 1 cells but not in animals that had higher frequencies of T H 1 cells, despite comparable vaccine-induced humoral and CD8 T cell immunity in both groups. An RNA transcriptome signature in blood at 7 days after priming immunization from one study was associated with induction of fewer T H 1-type CD4 cells and enhanced protection. These results demonstrate that high and persisting frequencies of HIV vaccine–induced T H 1-biased CD4 T cells in the intestinal and genital mucosa can mitigate beneficial effects of protective antibodies and CD8 T cells, highlighting a critical role of priming immunization and vaccine adjuvants in modulating HIV vaccine efficacy.
... Animals were confirmed negative for SIV and simian T cell lymphotropic virus (STLV) and were tested for the expression of Mamu-A*01, -B*08, and -B*17 alleles. TRIM5α genotyping was conducted as previously described (53). Animals were assigned to one of the immunization groups shown in Figure 1. ...
Article
Full-text available
Antibodies and cytotoxic T cells represent 2 arms of host defense against pathogens. We hypothesized that vaccines that induce both high-magnitude CD8+ T cell responses and antibody responses might confer enhanced protection against HIV. To test this hypothesis, we immunized 3 groups of nonhuman primates: (a) Group 1, which includes sequential immunization regimen involving heterologous viral vectors (HVVs) comprising vesicular stomatitis virus, vaccinia virus, and adenovirus serotype 5-expressing SIVmac239 Gag; (b) Group 2, which includes immunization with a clade C HIV-1 envelope (Env) gp140 protein adjuvanted with nanoparticles containing a TLR7/8 agonist (3M-052); and (c) Group 3, which includes a combination of both regimens. Immunization with HVVs induced very high-magnitude Gag-specific CD8+ T cell responses in blood and tissue-resident CD8+ memory T cells in vaginal mucosa. Immunization with 3M-052 adjuvanted Env protein induced robust and persistent antibody responses and long-lasting innate responses. Despite similar antibody titers in Groups 2 and 3, there was enhanced protection in the younger animals in Group 3, against intravaginal infection with a heterologous SHIV strain. This protection correlated with the magnitude of the serum and vaginal Env-specific antibody titers on the day of challenge. Thus, vaccination strategies that induce both CD8+ T cell and antibody responses can confer enhanced protection against infection.
Article
Cyclophilins (Cyps) are a subgroup of peptidyl-prolyl cis-trans isomerases (PPIases) that contain a highly conserved domain of PPIases. Sixteen Cyps have been identified in humans, among which the functions of five classical Cyp subtypes (CypA, B, C, D, and 40) have been studied in more detail. Cyps are widely expressed in almost all human tissues and are involved in several intracellular signaling pathways such as oxidative stress, mitochondrial dysfunction, cell migration, and apoptosis. Several studies have also demonstrated that Cyps play an important role in the development of cardiovascular diseases, neurodegeneration, cancer, and other diseases. However, as regulators of intercellular communication, Cyps have increasingly attracted attention as a result of their implications in viral infection. The specific motifs of Cyps can be targeted by viral proteins and thus promote either a viral infection or an antiviral response. This review highlights the present understanding of Cyps in viral infection and immune response. These effects will facilitate revealing the molecular mechanisms of several diseases induced by viruses and may provide novel insight into the development of corresponding drug-based treatment methods.
Article
Full-text available
There is currently a need for proxy measures of the HIV rebound competent reservoir (RCR) that can predict viral rebound after combined antiretroviral treatment (cART) interruption. In this study, macaques infected with a barcoded SIVmac239 virus received cART beginning between 4- and 27-days post-infection, leading to the establishment of different levels of viral dissemination and persistence. Later treatment initiation led to higher SIV DNA levels maintained during treatment, which was significantly associated with an increased frequency of SIV reactivation and production of progeny capable of causing rebound viremia following treatment interruption. However, a 100-fold increase in SIV DNA in PBMCs was associated with only a 2-fold increase in the frequency of reactivation. These data suggest that the RCR can be established soon after infection, and that a large fraction of persistent viral DNA that accumulates after this time makes relatively little contribution to viral rebound.
Article
The macaque simian or simian/human immunodeficiency virus (SIV/SHIV)challenge model has been widely used to inform and guide human vaccine trials. Substantial advances have been made recently in the application of repeated-low-dose challenge (RLD)approach to assess SIV/SHIV vaccine efficacies (VE). Some candidate HIV vaccines have shown protective effects in preclinical studies using the macaque SIV/SHIV model but the model's true predictive value for screening potential HIV vaccine candidates needs to be evaluated further. Here, we review key parameters used in the RLD approach and discuss their relevance for evaluating VE to improve preclinical studies of candidate HIV vaccines.
Article
Full-text available
We developed a method of simultaneous vaccination with DNA and protein resulting in robust and durable cellular and humoral immune responses with efficient dissemination to mucosal sites and protection against simian immunodeficiency virus (SIV) infection. To further optimize the DNA-protein coimmunization regimen, we tested a SIVmac251-based vaccine formulated with either of two Toll-like receptor 4 (TLR4) ligand-based liposomal adjuvant formulations (TLR4 plus TLR7 [TLR4+7] or TLR4 plus QS21 [TLR4+QS21]) in macaques. Although both vaccines induced humoral responses of similar magnitudes, they differed in their functional quality, including broader neutralizing activity and effector functions in the TLR4+7 group. Upon repeated heterologous SIVsmE660 challenge, a trend of delayed viral acquisition was found in vaccinees compared to controls, which reached statistical significance in animals with the TRIM-5α-resistant (TRIM-5α R) allele. Vaccinees were preferentially infected by an SIVsmE660 transmitted/founder virus carrying neutralizationresistant A/K mutations at residues 45 and 47 in Env, demonstrating a strong vaccine- induced sieve effect. In addition, the delay in virus acquisition directly correlated with SIVsmE660-specific neutralizing antibodies. The presence of mucosal V1V2 IgG binding antibodies correlated with a significantly decreased risk of virus acquisition in both TRIM-5α R and TRIM-5α-moderate/sensitive (TRIM-5α M/S) animals, although this vaccine effect was more prominent in animals with the TRIM-5α R allele. These data support the combined contribution of immune responses and genetic background to vaccine efficacy. Humoral responses targeting V2 and SIV-specific T cell responses correlated with viremia control. In conclusion, the combination of DNA and gp120 Env protein vaccine regimens using two different adjuvants induced durable and potent cellular and humoral responses contributing to a lower risk of infection by heterologous SIV challenge.
Article
Full-text available
The RV144 vaccine trial in Thailand demonstrated that an HIV vaccine could prevent infection in humans and highlights the importance of understanding protective immunity against HIV. We used a nonhuman primate model to define immune and genetic mechanisms of protection against mucosal infection by the simian immunodeficiency virus (SIV). A plasmid DNA prime/recombinant adenovirus serotype 5 (rAd5) boost vaccine regimen was evaluated for its ability to protect monkeys from infection by SIVmac251 or SIVsmE660 isolates after repeat intrarectal challenges. Although this prime-boost vaccine regimen failed to protect against SIVmac251 infection, 50% of vaccinated monkeys were protected from infection with SIVsmE660. Among SIVsmE660-infected animals, there was about a one-log reduction in peak plasma virus RNA in monkeys expressing the major histocompatibility complex class I allele Mamu-A*01, implicating cytotoxic T lymphocytes in the control of SIV replication once infection is established. Among Mamu-A*01-negative monkeys challenged with SIVsmE660, no CD8(+) T cell response or innate immune response was associated with protection against virus acquisition. However, low levels of neutralizing antibodies and an envelope-specific CD4(+) T cell response were associated with vaccine protection in these monkeys. Moreover, monkeys that expressed two TRIM5 alleles that restrict SIV replication were more likely to be protected from infection than monkeys that expressed at least one permissive TRIM5 allele. This study begins to elucidate the mechanisms of vaccine protection against immunodeficiency viruses and highlights the need to analyze these immune and genetic correlates of protection in future trials of HIV vaccine strategies.
Article
Full-text available
TRIM5 is a RING domain-E3 ubiquitin ligase that restricts infection by human immunodeficiency virus (HIV)-1 and other retroviruses immediately following virus invasion of the target cell cytoplasm. Antiviral potency correlates with TRIM5 avidity for the retrovirion capsid lattice and several reports indicate that TRIM5 has a role in signal transduction, but the precise mechanism of restriction is unknown. Here we demonstrate that TRIM5 promotes innate immune signalling and that this activity is amplified by retroviral infection and interaction with the capsid lattice. Acting with the heterodimeric, ubiquitin-conjugating enzyme UBC13-UEV1A (also known as UBE2N-UBE2V1), TRIM5 catalyses the synthesis of unattached K63-linked ubiquitin chains that activate the TAK1 (also known as MAP3K7) kinase complex and stimulate AP-1 and NFκB signalling. Interaction with the HIV-1 capsid lattice greatly enhances the UBC13-UEV1A-dependent E3 activity of TRIM5 and challenge with retroviruses induces the transcription of AP-1 and NF-κB-dependent factors with a magnitude that tracks with TRIM5 avidity for the invading capsid. Finally, TAK1 and UBC13-UEV1A contribute to capsid-specific restriction by TRIM5. Thus, the retroviral restriction factor TRIM5 has two additional activities that are linked to restriction: it constitutively promotes innate immune signalling and it acts as a pattern recognition receptor specific for the retrovirus capsid lattice.
Article
Full-text available
Rapid evolution and high intrahost sequence diversity are hallmarks of human and simian immunodeficiency virus (HIV/SIV) infection. Minor viral variants have important implications for drug resistance, receptor tropism, and immune evasion. Here, we used ultradeep pyrosequencing to sequence complete HIV/SIV genomes, detecting variants present at a frequency as low as 1%. This approach provides a more complete characterization of the viral population than is possible with conventional methods, revealing low-level drug resistance and detecting previously hidden changes in the viral population. While this work applies pyrosequencing to immunodeficiency viruses, this approach could be applied to virtually any viral pathogen.
Article
Full-text available
Simian immunodeficiency viruses of sooty mangabeys (SIVsm) are the source of multiple, successful cross-species transmissions, having given rise to HIV-2 in humans, SIVmac in rhesus macaques, and SIVstm in stump-tailed macaques. Cellular assays and phylogenetic comparisons indirectly support a role for TRIM5alpha, the product of the TRIM5 gene, in suppressing interspecies transmission and emergence of retroviruses in nature. Here, we investigate the in vivo role of TRIM5 directly, focusing on transmission of primate immunodeficiency viruses between outbred primate hosts. Specifically, we retrospectively analyzed experimental cross-species transmission of SIVsm in two cohorts of rhesus macaques and found a significant effect of TRIM5 genotype on viral replication levels. The effect was especially pronounced in a cohort of animals infected with SIVsmE543-3, where TRIM5 genotype correlated with approximately 100-fold to 1,000-fold differences in viral replication levels. Surprisingly, transmission occurred even in individuals bearing restrictive TRIM5 genotypes, resulting in attenuation of replication rather than an outright block to infection. In cell-culture assays, the same TRIM5 alleles associated with viral suppression in vivo blocked infectivity of two SIVsm strains, but not the macaque-adapted strain SIVmac239. Adaptations appeared in the viral capsid in animals with restrictive TRIM5 genotypes, and similar adaptations coincide with emergence of SIVmac in captive macaques in the 1970s. Thus, host TRIM5 can suppress viral replication in vivo, exerting selective pressure during the initial stages of cross-species transmission.
Article
Full-text available
An effective human immunodeficiency virus (HIV) vaccine will likely need to reduce mucosal transmission and, if infection occurs, control virus replication. To determine whether our best simian immunodeficiency virus (SIV) vaccine can achieve these lofty goals, we vaccinated eight Indian rhesus macaques with SIVmac239Delta nef and challenged them intrarectally (i.r.) with repeated low doses of the pathogenic heterologous swarm isolate SIVsmE660. We detected a significant reduction in acquisition of SIVsmE660 in comparison to that for naïve controls (log rank test; P = 0.023). After 10 mucosal challenges, we detected replication of the challenge strain in only five of the eight vaccinated animals. In contrast, seven of the eight control animals became infected with SIVsmE660 after these 10 challenges. Additionally, the SIVsmE660-infected vaccinated animals controlled peak acute virus replication significantly better than did the naïve controls (Mann-Whitney U test; P = 0.038). Four of the five SIVsmE660 vaccinees rapidly brought virus replication under control by week 4 postinfection. Unfortunately, two of these four vaccinated animals lost control of virus replication during the chronic phase of infection. Bulk sequence analysis of the circulating viruses in these animals indicated that recombination had occurred between the vaccine and challenge strains and likely contributed to the increased virus replication in these animals. Overall, our results suggest that a well-designed HIV vaccine might both reduce the rate of acquisition and control viral replication.
Article
Full-text available
The cytoplasmic TRIM5alpha proteins of certain mammalian lineages efficiently recognize the incoming capsids of particular retroviruses and potently restrict infection in a species-specific manner. Successful retroviruses have evolved capsids that are less efficiently recognized by the TRIM5alpha proteins of the natural hosts. To address whether TRIM5alpha contributes to the outcome of retroviral infection in a susceptible host species, we investigated the impact of TRIM5 polymorphisms in rhesus monkeys on the course of a simian immunodeficiency virus (SIV) infection. Full-length TRIM5alpha cDNAs were derived from each of 79 outbred monkeys and sequenced. Associations were explored between the expression of particular TRIM5 alleles and both the permissiveness of cells to SIV infection in vitro and clinical sequelae of SIV infection in vivo. Natural variation in the TRIM5alpha B30.2(SPRY) domain influenced the efficiency of SIVmac capsid binding and the in vitro susceptibility of cells from the monkeys to SIVmac infection. We also show the importance in vivo of the interaction of SIVmac with different allelic forms of TRIM5, demonstrating that particular alleles are associated with as much as 1.3 median log difference in set-point viral loads in SIVmac-infected rhesus monkeys. Moreover, these allelic forms of TRIM5 were associated with the extent of loss of central memory (CM) CD4+ T cells and the rate of progression to AIDS in the infected monkeys. These findings demonstrate a central role for TRIM5alpha in limiting the replication of an immunodeficiency virus infection in a primate host.
Article
Full-text available
Recent findings have highlighted roles played by innate cellular factors in restricting intracellular viral replication. In this review, we discuss in brief the activities of apolipoprotein B mRNA-editing enzyme 3G (APOBEC3G), bone marrow stromal cell antigen 2 (BST-2), cyclophilin A, tripartite motif protein 5 alpha (Trim5alpha), and cellular microRNAs as examples of host restriction factors that target HIV-1. We point to countermeasures encoded by HIV-1 for moderating the potency of these cellular restriction functions.
Article
Full-text available
We recently developed a novel strategy to identify transmitted HIV-1 genomes in acutely infected humans using single-genome amplification and a model of random virus evolution. Here, we used this approach to determine the molecular features of simian immunodeficiency virus (SIV) transmission in 18 experimentally infected Indian rhesus macaques. Animals were inoculated intrarectally (i.r.) or intravenously (i.v.) with stocks of SIVmac251 or SIVsmE660 that exhibited sequence diversity typical of early-chronic HIV-1 infection. 987 full-length SIV env sequences (median of 48 per animal) were determined from plasma virion RNA 1-5 wk after infection. i.r. inoculation was followed by productive infection by one or a few viruses (median 1; range 1-5) that diversified randomly with near starlike phylogeny and a Poisson distribution of mutations. Consensus viral sequences from ramp-up and peak viremia were identical to viruses found in the inocula or differed from them by only one or a few nucleotides, providing direct evidence that early plasma viral sequences coalesce to transmitted/founder viruses. i.v. infection was >2,000-fold more efficient than i.r. infection, and viruses transmitted by either route represented the full genetic spectra of the inocula. These findings identify key similarities in mucosal transmission and early diversification between SIV and HIV-1, and thus validate the SIV-macaque mucosal infection model for HIV-1 vaccine and microbicide research.
Article
Full-text available
All human immunodeficiency virus (HIV) vaccine efficacy trials to date have ended in failure. Structural features of the Env glycoprotein and its enormous variability have frustrated efforts to induce broadly reactive neutralizing antibodies. To explore the extent to which vaccine-induced cellular immune responses, in the absence of neutralizing antibodies, can control replication of a heterologous, mucosal viral challenge, we vaccinated eight macaques with a DNA/Ad5 regimen expressing all of the proteins of SIVmac239 except Env. Vaccinees mounted high-frequency T-cell responses against 11 to 34 epitopes. We challenged the vaccinees and eight naïve animals with the heterologous biological isolate SIVsmE660, using a regimen intended to mimic typical HIV exposures resulting in infection. Viral loads in the vaccinees were significantly less at both the peak (1.9-log reduction; P < 0.03) and at the set point (2.6-log reduction; P < 0.006) than those in control naïve animals. Five of eight vaccinated macaques controlled acute peak viral replication to less than 80,000 viral RNA (vRNA) copy eq/ml and to less than 100 vRNA copy eq/ml in the chronic phase. Our results demonstrate that broad vaccine-induced cellular immune responses can effectively control replication of a pathogenic, heterologous AIDS virus, suggesting that T-cell-based vaccines may have greater potential than previously appreciated.
Article
Retroviruses have evolved effective strategies to evade the host immune response, such as high variability and latent infection. In addition, primate lentiviruses, such as HIV-1, have acquired several "accessory" genes that antagonize antiviral host restriction factors and facilitate viral immune evasion, thereby allowing continuous and efficient viral replication despite apparently strong innate and acquired immune responses. Here, I summarize some of our current knowledge on the acquisition and function of the viral vif, vpr, vpu, and nef genes, with a particular focus on the evolution and specific properties of pandemic HIV-1 strains that may contribute to their efficient spread and high virulence.