You are on page 1of 14

Article

HIV-1-Infected CD4+ T Cells Facilitate Latent


Infection of Resting CD4+ T Cells through Cell-Cell
Contact
Graphical Abstract Authors
Luis M. Agosto, Melissa B. Herring,
Walther Mothes, Andrew J. Henderson

Correspondence
agosto@bu.edu (L.M.A.),
andrew.henderson@bmc.org (A.J.H.)

In Brief
Agosto et al. demonstrate that HIV-1 cell-
to-cell transmission between T cells
mediates the generation of latent
infection of resting CD4+ T cells. This
mechanism for the formation of latent
infection may be more difficult to reverse
and poses a potential hurdle for the
development of latency-reversing
therapies.

Highlights
d Developed in vitro model of HIV-1 latency incorporating HIV
cell-to-cell transmission

d HIV cell-to-cell transmission infects resting T cells without


robust T cell activation

d Proviruses generated are more difficult to induce, posing a


barrier to HIV eradication

Agosto et al., 2018, Cell Reports 24, 2088–2100


August 21, 2018 ª 2018 The Author(s).
https://doi.org/10.1016/j.celrep.2018.07.079
Cell Reports

Article

HIV-1-Infected CD4+ T Cells Facilitate Latent


Infection of Resting CD4+ T Cells
through Cell-Cell Contact
Luis M. Agosto,1,4,* Melissa B. Herring,1,2 Walther Mothes,3 and Andrew J. Henderson1,*
1Department of Medicine, Section of Infectious Diseases, Boston University Medical Center, Boston, MA, USA
2St. Georges University of London, London, UK
3Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
4Lead Contact

*Correspondence: agosto@bu.edu (L.M.A.), andrew.henderson@bmc.org (A.J.H.)


https://doi.org/10.1016/j.celrep.2018.07.079

SUMMARY cell-free HIV, such as antiviral factors, neutralizing antibodies,


some anti-retroviral therapies, and poor viral fitness (Agosto
HIV-1 is transmitted between T cells through the et al., 2015b; Brandenberg et al., 2014; Gupta et al., 1989; Jolly
release of cell-free particles and through cell-cell et al., 2010; Mathez et al., 1993; Sigal et al., 2011; Zhong et al.,
contact. Cell-to-cell transmission is more efficient 2013a). However, despite the efficiency of cell-to-cell HIV-1
than cell-free virus transmission, mediates resis- transmission, it remains unclear whether this process contrib-
tance to immune responses, and facilitates the utes to the pathogenesis of HIV.
The most significant barrier for the eradication of HIV-1 infec-
spread of virus among T cells. However, whether
tion is the existence of a small pool of latently infected cells that
HIV cell-to-cell transmission influences the establish-
survive despite long-term suppression of viral replication by
ment of HIV-1 latency has not been carefully highly active anti-retroviral therapy (HAART) (Finzi et al., 1997;
explored. We developed an HIV-1 latency model Ruelas and Greene, 2013; Strain et al., 2003). Following treat-
based on the transmission of HIV-1 directly to resting ment interruption, this pool of cells contributes to the re-emer-
CD4+ T cells by cell-cell contact. This model recapit- gence of viremia (Chun et al., 1997b; Davey et al., 1999; Wong
ulates the spread of HIV-1 in T-cell-dense anatomical et al., 1997). The reservoir of latently infected T cells is estab-
compartments. We demonstrate that productively lished within 3–10 days after exposure to virus and prior to the
infected activated CD4+ T cells transmit HIV-1 to detection of viremia (Chun et al., 1998; Whitney et al., 2014).
resting CD4+ T cells in a cell-contact-dependent The vast majority of these latently infected cells are composed
manner. However, proviruses generated in this of resting memory CD4+ T cells that have a long half-life and
are maintained by homeostatic proliferation without the induc-
fashion are more difficult to induce compared to pro-
tion of viral replication (Brenchley et al., 2004; Chomont et al.,
viruses generated by cell-free infection, suggesting
2009; Chun et al., 1997a; Douek et al., 2002; Maldarelli et al.,
that cell-to-cell transmission influences the estab- 2014; Ostrowski et al., 1999; Wagner et al., 2014). Significant
lishment and maintenance of latent infection in progress has been made toward understanding the molecular
resting CD4+ T cells. mechanisms that regulate proviral transcription and latency in
T cells. Latently infected T cells are characterized by inefficient
INTRODUCTION proviral transcription due to insufficient expression and binding
of cellular transcription factors to the proviral promoter, the inef-
HIV-1 spreads by both cell-free virions and by cell-contact- ficient elongation of transcription, and a closed chromatin envi-
dependent transmission between CD4+ T cells (Jolly et al., ronment (Agosto et al., 2015a; Mbonye and Karn, 2017; Ruelas
2004; Sattentau, 2008). The latter form of viral transmission is and Greene, 2013). Despite the knowledge of these molecular
several orders of magnitude more efficient than the spread of mechanisms that regulate latent infection, it remains unclear
cell-free particles (Chen et al., 2007; Zhong et al., 2013b). Two how the pool of latently infected resting CD4+ T cells is initially
important features may account for the high efficiency of cell- generated.
to-cell viral transmission. First, cell contacts promote the forma- Early studies suggested that cell activation, as defined by cell
tion of virally induced junctions, or virological synapses, between cycle progression beyond G1b, is not only required for proviral
CD4+ T cells, which concentrate large numbers of particles at production but also for efficient reverse transcription and pro-
the site of cell-cell contact (Johnson and Huber, 2002; Jolly viral integration (Bukrinsky et al., 1991; Korin and Zack, 1998;
et al., 2004; Phillips, 1994). This process delivers a high MOI to Stevenson et al., 1990; Sun and Clark, 1999; Unutmaz et al.,
the target cell (Del Portillo et al., 2011; Duncan et al., 2014; Zhong 1999; Zack et al., 1992). This led to the hypothesis that latently
et al., 2013a). Second, cell contact helps to overcome cellular infected cells are generated as activated HIV-infected CD4+
and environmental barriers that normally prevent infection by T cells return to a resting state (Ruelas and Greene, 2013).

2088 Cell Reports 24, 2088–2100, August 21, 2018 ª 2018 The Author(s).
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Wash Alu-PCR Targets
+SQV

48-72h aCD4 24h 72h


aCD4 rCD4
FACS for HIV+
+RAL Targets
Co-Culture +Stimulation
HIV 1 donor: 1 target 24h rCD4

1K +EFV No Drug 100


800 80

Frequency (%)
0.5% 23.3+/-2.6%

Count
FSC

600 60
400 40
200 20
0 0 1 2 3 4 0
10 10 10 10 10 100 101 102 10
3
10
4
0 102 103 104 105 Targets Viability
HIV-Gag CD69/HLA-DR/CD25
Resting cells
Activated cells
Unstained

Figure 1. In Vitro Model of HIV-1 Cell-to-Cell Transmission


See text and Experimental Procedures for details. A flow cytometry profile for HIV Gag expression in activated CD4+ T cells in the presence or absence of EFV is
shown. The average percent of HIV-1 Gag expression ± SD from 8 experiments is indicated. A flow cytometry profile of activation marker expression among
purified resting CD4+ T cells and activated CD4+ T cells is shown. We estimated the percent of target resting CD4+ T cells and their viability at the end of the co-
culture period.

An alternative mechanism for the formation of latently infected In the present study, we developed a model of HIV-1 latency
CD4+ T cells is that resting cells are directly infected. Although that incorporates HIV-1 cell-to-cell transmission directly to
direct infection of resting CD4+ T cells has been estimated to resting CD4+ T cells. With this model, we demonstrate that
be 10-fold less efficient compared to activated cells, the pro- productively infected CD4+ T cells mediate latent infection
cess can be successfully completed but with slower kinetics in target resting CD4+ T cells in a cell-contact-mediated
than in activated cells (Agosto et al., 2007; Lassen et al., fashion. Infection via this mechanism generates a population
2012; Plesa et al., 2007; Swiggard et al., 2005; Vatakis et al., of cells harboring latent proviruses that are more difficult to
2007). Resting CD4+ T cells represent a large proportion of reactivate. Taken together, our studies support a mechanism
the total population of CD4+ T cells, and because these cells for the generation of latently infected cells through direct infec-
are already at a cellular state that would favor latent infection, tion of resting CD4+ T cells and provide evidence for the
it is possible that directly infected resting cells may represent importance of T cell-T cell interactions in the pathogenesis
a significant proportion of the latent reservoir. Understanding of HIV.
the requirements for infection of these cells is therefore impor-
tant for better understanding HIV-1 pathogenesis (Zack et al., RESULTS
2013). Several factors could influence the susceptibility of
resting CD4+ T cells to HIV-1 infection without resulting in full Modeling HIV-1 Cell-to-Cell Transmission to Resting
activation of the cell and progression through the cell cycle. CD4+ T Cells
These factors may include the cytokine environment in tissues To test whether productively infected activated CD4+ T cells
and cell-cell interactions (Eckstein et al., 2001; Kinter et al., transmit HIV-1 to resting CD4+ T cells, we employed a co-cul-
2003; Nishimura et al., 2005; Saleh et al., 2007; Shen et al., ture approach in which activated CD4+ T cells productively in-
2013; Zhang et al., 1999). Indeed, as previously shown by fected with HIV-1 were co-cultured with autologous target
Evans et al. (2013) and Kumar et al. (2015), cell-cell interac- resting CD4+ T cells (Figure 1). 20%–30% of infected activated
tions, such as those between antigen-presenting cells and cells expressed HIV-1 as assessed by intracellular HIV-1 Gag
CD4+ T cells, can mediate latent infection in CD4+ T cells. (Figure 1). Resting CD4+ T cells were purified from total CD4+
Thus, it is likely that cell-to-cell transmission between CD4+ T cells by depleting cells expressing CD25, CD69, and HLA-
T cells may facilitate the establishment of latency by increasing DR (Figure 1); absence of these markers corresponds to
the likelihood of direct infection of resting CD4+ T cells. T cells at the quiescent and/or resting G0/G1a stage of the cell

Cell Reports 24, 2088–2100, August 21, 2018 2089


cycle (Agosto et al., 2009; Korin and Zack, 1998). Resting CD4+ Productively Infected Activated CD4+ T Cells Form
T cells were stained with eFluor670 prior to co-culture to allow Contacts with Resting CD4+ T Cells
us to track them at the end of the incubation period. The cells We examined whether infection of target resting CD4+ T cells
were co-cultured for 24 hr and were then treated with the HIV following co-culture with productively infected activated
protease inhibitor saquinavir (SQV). Treatment with SQV pre- CD4+ T cells was dependent on cell-cell contact. To test this,
vents maturation of viral particles released from infected acti- resting CD4+ T cells and productively infected activated
vated CD4+ T cells, thus preventing the spread of de-novo- CD4+ T cells were co-cultured as described above or sepa-
assembled virus. This treatment allowed us to examine the rated by a 0.4-mm membrane Transwell (Figure 4A). We then
transfer of HIV-1 that occurs specifically within the first 24 hr assessed the level of HIVNL4-3 integration by Alu-PCR after
of co-culture (Agosto et al., 2014; Titanji et al., 2013). Once 72 hr of culture. Whereas HIV integration is detected in resting
SQV was added, the culture was incubated for 3 days to allow cells after co-culture, we were unable to detect proviral DNA
time for HIV-1 to reverse transcribe and integrate in the target PCR signal in Transwell cultures above the background signal
cells. This extended incubation is necessary as the kinetics of from EFV-treated cultures (Figure 4B). To confirm that viral par-
reverse transcription and integration are slower in resting cells ticles released by productively infected CD4+ T cells cross the
(Plesa et al., 2007). At the end of the co-culture period, we 0.4-mm membrane, we plated the HIV indicator cell line TZMbl
confirmed that the percent of target resting CD4+ T cells was in the lower well. This indicator cell line, which is highly suscep-
maintained at approximately 50% and that their viability was tible to HIV-1, is transduced with an HIV Tat-responsive re-
maintained at approximately 80% (Figure 1). At this point, target porter construct that expresses firefly luciferase upon HIV
cells were separated from infected activated cells by cell sorting infection. We detected a significant luciferase signal above
and assayed for proviral DNA by Alu-PCR (Figure 2). This PCR background, confirming that viral particles crossed the mem-
approach, previously described in detail (Liszewski et al., brane and successfully infected cells in the bottom well (Fig-
2009), is based on an initial amplification of integrated HIV using ure 4C). Taken together, these data suggest that T cell-T cell
a reverse primer specific for the HIV gag gene and a forward contact more efficiently facilitates the transfer of HIV-1 from
primer specific for human Alu. A second quantitative nested productively infected cells to resting CD4+ T cells compared
PCR amplification is performed with the reaction product of to the release of cell-free particles in our co-culture system.
the first amplification as a template, using HIV-specific primers This observation is consistent with published work conducted
recognizing the R-U5 region and a TaqMan probe (Figure 2B). with activated CD4+ T cells and cell lines indicating that cell-
Signals from experimental samples were plotted against a poly- cell-contact-mediated HIV-1 transmission is largely responsible
clonal integration standard to determine the number of proviral for the spread of HIV in culture (Chen et al., 2007; Sourisseau
copies per cell. This Alu-PCR technique has a detection sensi- et al., 2007; Zhong et al., 2013a).
tivity of a single HIV integration event per 10,000 cells (Agosto To directly assess whether productively infected CD4+ T cells
et al., 2007). Treatment of cultures with the anti-retroviral drug interact with resting CD4+ T cells and to gain insight as to what
efavirenz (EFV) provides the background signal for both qPCR- modes of viral transmission are involved, we visualized cell-cell
and flow cytometry-based experiments. We detected proviral contacts by confocal microscopy. We infected activated CD4+
DNA in target resting CD4+ T cells after co-culture with produc- T cells with HIVNL4-3-Gag-GFP and co-cultured them with resting
tively infected activated CD4+ T cells compared to co-cultures CD4+ T cells for 2 hr prior to confocal microscopy (Figure 4D).
treated with EFV (Figure 2C). The amount of proviral DNA corre- Because HIVNL4-3-Gag-GFP particles are fluorescent, we deter-
sponded to an infection rate of 1%–15% of resting CD4+ T cells. mined the frequency of HIV-1 Gag-GFP fluorescence associated
Importantly, we detected integration in target resting CD4+ with target cells in cell-cell contacts (Figure 4E) compared to the
T cells when using both X4-tropic HIV-1 (HIVNL4-3) and the frequency of fluorescence in resting CD4+ T cells that were not
more physiologically relevant R5-tropic HIV-1 (HIVREJO). This is engaged in cell-cell contacts (Figure 4F). We observed that
consistent with expression of CXCR4 and CCR5 expression target resting CD4+ T cells were more likely associated with
on resting CD4+ T cells (Figure S1; Dai et al., 2009; Tabler HIV-1 Gag-GFP fluorescence when conjugated with activated
et al., 2014; Zerbato et al., 2016). CD4+ T cells compared to cells not in contact with activated
Cell-cell interactions and the culture microenvironment can CD4+ T cells (Figure 4G).
affect the phenotype and function of cells. We wanted to deter- Formation of cell-cell contacts is a dynamic process often re-
mine whether the co-culture microenvironment leads to cell sulting in the polarization of HIV assembly to the sites of cell-cell
activation of the resting CD4+ T cells. We monitored the expres- contact. This polarization of viral assembly is a defining charac-
sion of the activation markers CD69, HLA-DR, and CD25 among teristic of virological synapses (Chen et al., 2007; Jolly et al.,
the target resting CD4+ T cell population at various time points 2004). Therefore, we monitored the formation of cell-cell conju-
post-co-culture by flow cytometry. We observed that, although gates using live-cell imaging, quantified the duration of the con-
target resting CD4+ T cells did not demonstrate an activated jugates, and surveyed the frequency of HIV Gag-GFP polariza-
phenotype throughout the co-culture when compared to acti- tion to the site of cell-cell contact (Figure 5; Videos S1, S2, and
vated CD4+ T cells, there was a modest but statistically signif- S3). We observed that productively infected activated CD4+
icant induction of T cell activation marker expression (Figure 3). T cells formed conjugates with resting CD4+ T cells and could
These data suggest that weak cell signaling is involved in the observe the transfer of particles in some instances, although
process of HIV-1 cell-to-cell transmission to target resting we lack the resolution to see the transfer in all cell-cell conju-
CD4+ T cells. gates. Approximately 26% of conjugates resulted in the

2090 Cell Reports 24, 2088–2100, August 21, 2018


A

B C

Figure 2. HIV-Producing T Cells Transmit HIV-1 Directly to Resting CD4+ T Cells


(A) After co-culture, target resting CD4+ T cells were sorted to assess the level of HIV-1 integration within this population. The flow cytometry plots show the
sorting strategy.
(B) Simplified diagram of the Alu-PCR strategy to specifically detect HIV integration. Shown are the relative primer locations.
(C) HIV-1 integration was assessed in co-cultures infected with HIVNL4-3 (X4) and HIVREJO (R5) by Alu-PCR. Parallel control co-cultures were treated with EFV.
Symbols represent independent blood donors. Integration was measured 5 times for each sample. **p < 0.01.
Error bars represent the SD of the mean.

accumulation of HIV Gag at the site of cell-cell contact. Conju- vations support our hypothesis that cell-cell contacts facilitate
gates that resulted in the polarization of HIV Gag-GFP were the transfer of HIV-1 to resting CD4+ T cells in co-cultures.
generally longer lived (average 33 min) compared to conjugates Several modes of cell-contact-dependent viral transmission
that did not result in polarization (average 23 min). These obser- between cells have been described. In the case of HIV-1, virus

Cell Reports 24, 2088–2100, August 21, 2018 2091


A B We detected a significant amount of HIV-1 Gag expression in

CD25/CD69/HLA-DR (MFI)
10 4
target cells after stimulation of the co-culture compared to unsti-
mulated co-cultures (Figure 7A). The presence of inducible pro-
10 3 viral HIV-1 expression supports the conclusion that co-culture
Count

* * * with productively infected activated CD4+ T cells leads to the


2
10 generation of latent infection in resting CD4+ T cells.
The ratio of the percent HIV-1 Gag-positive cells to the percent
10 1 cells with integrated HIV-1 provides insight into the subset of
2 3 4 5
0 10 10 10 10
cells that have reversible latent infection. For the example shown
D ore e
C

D y1 e
2 CC

D y2 e
C
ay ne

ed
iv C
ef p

D lon
C

A 4C
B ty

at
CD69/HLA-DR/CD25 l

l
o

A
in Figure 7A, approximately 1.07% of the target resting CD4+
Is

ct
1

4
a

a
D

D
ay

ay

ay
Isotype Day 2
Day 0
T cells express HIV-1 Gag once the percent background expres-
Day 4
Day 1 Activated sion from unstimulated target cells is subtracted. This percent-
age reflects the proportion of cells with inducible proviruses.
Figure 3. Co-culture with Productively Infected Activated CD4+ Before stimulation, we estimated that 4.4% of the total target
T Cells Does Not Induce Robust Activation of Resting CD4+ T Cells resting CD4+ T cell population (or 0.044 proviruses/cell) carried
(A) We assessed the level of T cell activation among target resting CD4+ T cells integrated HIV-1 as determined by Alu-PCR. Therefore, we
at various time points post-co-culture based on the expression of CD69, HLA- calculated, by dividing the percent of HIV-Gag-positive cells by
DR, and CD25. The levels of activation based on the expression of activation
the percent of cells carrying proviruses, that approximately
markers (mean fluorescence index) were compared to activated CD4+ T cells
treated with PHA, IL-2, and IL-7. 24% of infected cells could be induced to produce HIV-1 upon
(B) Combined data from 3 individual experiments from three different blood stimulation. Based on four independent experiments, an average
donors. Cells cultured alone or in co-culture (CC) with infected CD4+ T cells are of 2.6% of resting cells carried proviruses after co-culture infec-
shown. The horizontal line represents the average of each population. *p < tion (Figure 7D) and an average of 21% of proviruses were induc-
0.05. ible (Figure 7E). To gain further insight into the relative efficiency
of the induction of proviruses in resting CD4+ T cells after cell-to-
cell transmission, we tested the induction of latent infection
cell-to-cell transmission can be mediated by virological synap-
generated by spinoculating resting CD4+ T cells with HIVNL4-3.
ses as previously stated. However, HIV-1 particles can also be
This model of HIV-1 latency involves the centrifugation of
transmitted without polarization. For example, HIV-1 particles
concentrated HIV-1 supernatant directly onto resting CD4+
produced by infected cells can be retained by infected cells
T cells (Swiggard et al., 2005). Because pure resting CD4+
and transferred to adjacent uninfected cells (Sherer et al.,
T cells are relatively resistant to HIV-1 infection compared to acti-
2010; Zhong et al., 2013b) or particles can be captured by unin-
vated T cells, delivering a larger amount of viral particles in-
fected cells and subsequently transferred to other uninfected
creases the proportion of resting cells that become infected
cells. We examined our co-cultures using microscopy to deter-
while preserving the resting nature of the cells (Agosto et al.,
mine whether there was evidence of alternative modes of viral
2007, 2009). Differences in the induction efficiency of proviruses
transmission. We detected both cell-cell contacts with polarized
between the co-culture model and the spinoculation model
HIV-1 assembly (Figure 6A) and cell contacts without polarized
would suggest that co-culture conditions could have a direct
virus (homogeneous cytoplasmic GFP staining and CD4
impact on the nature of latent infection in target resting CD4+
depleted; Figure 6B). We also found cell contacts between unin-
T cells. We infected resting CD4+ T cells from 4 blood donors
fected cells carrying viral particles and uninfected resting CD4+
by spinoculation and obtained HIV-1 expression in all donors
T cells (GFP punctae and CD4-positive; Figure 6C). This obser-
upon induction with a combination of anti-CD3/CD28 beads,
vation suggests that uninfected T cells carrying bound HIV could
IL-2, and IL-7 in the presence of raltegravir (Figure 7B). In the
contribute to the infection of resting CD4+ T cells. We observed
example shown in Figure 7B, 2.87% of cells expressed HIVNL4-3
that the frequency of GFP signal on resting CD4+ T cells in the
Gag (background from unstimulated cells subtracted), and about
context of polarized HIV was approximately 40%. Whereas
6.2% of the resting CD4+ T cells carried integrated HIV-1 (or
these images suggest that both polarized and non-polarized viral
0.062 proviruses/cell) prior to stimulation. Therefore, we found
transmission lead to the transfer of viral particles to resting CD4+
that approximately 46% of proviruses were induced upon stim-
T cells, single snapshots may not fully capture the dynamic pro-
ulation of the cells. From 5 experiments, an average of 13.5%
cess of viral transmission (Figure 6D).
of resting cells carried proviruses after spinoculation (Figure 7D)
and an average of 43% of proviruses were inducible (Figure 7E).
Latent Infection in the Target Cell Population Is The fraction of inducible proviruses after spinoculation
Inducible compared to co-culturing is statistically higher and suggests
Because we could detect HIV-1 integration among target resting that co-culture conditions contribute to the generation of provi-
CD4+ T cells after transmission from productively infected ruses more resistant to induction.
T cells, we tested whether proviral expression was inducible. To examine other more physiological modes of infection of
As described in Figure 1, co-cultures were treated with raltegra- resting CD4+ T cells, we also investigated the proportion of
vir and stimulated with a combination of anti-CD3/CD28 beads, inducible proviruses when resting CD4+ T cells were infected
interleukin-2 (IL-2), and IL-7. HIV-1NL4-3 expression was as- with concentrated HIVNL4-3 without spinoculation. To achieve
sessed by intracellular Gag staining and flow cytometry analysis. detectable HIV infection in resting CD4+ T cells without

2092 Cell Reports 24, 2088–2100, August 21, 2018


A B NS
C
10 0 1500 *

Infectivity (Luc RLU)


**

Proviruses/cell
-1
10
1000
10 -2
10 -3
500
10 -4
Co-culture Cell-free
10 -5 0
HIV-1 donor cells +EFV No Drug

re

l
FV

el
Target cells

tu

sw
+E

ul

an
C
o-

Tr
C
gag tat

D MA CA NC p6 gfp vif vpu


U3 R U5 U3 R U5
Δpol vpr env nef

rev

E Cell-Cell Contact F No Cell-Cell Contact G

rCD4T w/ GFP Signal (%)


100
*
80
60
P
40
20
0
T No Contact
Contact

Figure 4. HIV-1 Integrates in Resting CD4+ T Cells in a Cell-Contact-Dependent Manner


(A) A Transwell insert system was used to compare the relative efficiency of resting CD4+ T cell infection after co-culture with HIV-1-infected activated cells or
when the infected activated cells were separated.
(B) HIV-1 integration was measured by Alu-PCR in the target resting CD4+ T cells after co-culture or Transwell-based infection. Note that the co-culture dataset is
the same as Figure 1B for HIVNL4-3.
(C) To confirm that viral particles produced by infected activated CD4+ T cells can diffuse through the membrane, TZMbl cells were plated in the bottom well.
Infection was assessed by luciferase expression. Error bars represent the SD of 3 measurements of luciferase. A parallel control culture was treated with EFV.
(D) Diagram of the HIVNL4-3-Gag-GFP genome.
(E and F) Activated CD4+ T cells producing HIVNL4-3-Gag-GFP (green, P) were co-cultured with target resting CD4+ T cells pre-labeled with CMAC (blue, T) and
stained for CD4 (red). An example of a resting CD4+ T cell that carried HIV-1 Gag-GFP signal and was also in contact with an HIV-1-producer T cell is shown in (E).
An example of a resting CD4+ T cell that was not in contact with HIV-1-producer T cells but carried HIV-1 Gag-GFP signals is shown in (F). The scale bars in the
images represent 5 mm.
(G) The relative proportion of each is shown.
Error bars represent the SD of the mean. In (B), (C), and (G), *p < 0.05, **p < 0.01.

spinoculation, we used 5 times concentrated HIVNL4-3 compared cells suggests that reversing latent proviruses partly reflects how
to the spinoculation experiments. Under these conditions, we the infection was initially established.
observed inducible latent HIV in 3 out of 5 experiments (Fig-
ure 7C). From these experiments, we detected an average of DISCUSSION
5.6% of resting cells carrying proviruses (0.056 proviruses/cell;
Figure 7D). Importantly, this level of HIV-1 integration is compa- Cell-contact-mediated spread of HIV-1 has been estimated to
rable to the levels detected in co-culture experiments. On be at least an order of magnitude more efficient compared to
average, 31% of proviruses were inducible (Figure 7E). This pro- passive dissemination of particles through the extracellular
portion of inducible proviruses is intermediate between that of milieu. Thus, this form of viral transmission can have significant
proviruses generated by cell-to-cell transmission and proviruses effects on the pathogenesis of HIV. First, this mode of transmis-
generated by spinoculation. The observed differences in the abil- sion can mediate the spread of HIV-1 within T-cell-dense
ity to reactivate provirus in different models of infection of resting anatomical compartments (Law et al., 2016; Murooka et al.,

Cell Reports 24, 2088–2100, August 21, 2018 2093


A Figure 5. Contacts between Productively In-
fected CD4+ T Cells and Uninfected Resting
CD4+ T Cells Result in Polarization of HIV
Gag to the Site of Cell-Cell Contact
(A) Live-cell imaging was conducted to observe
conjugate formation between activated CD4+
T cells infected with HIVNL4-3-Gag-GFP (green) and
uninfected resting CD4+ T cells in real time. Images
were collected every 2.5 min. Shown are selected
time points when the transfer of particles was de-
tected (white arrows) and when polarization of HIV
Gag-GFP to the site of cell-cell contact was
detected (orange arrow). The full movie is provided
as Video S1. The scale bars in the images repre-
sent 5 mm.
(B) Cell-cell contacts were identified and scored
based on their duration. Contacts maintained for
least 5 min were scored. The duration of cell-cell
contacts that did not result in HIV Gag-GFP polari-
zation were compared to the duration of contacts
that resulted in polarization. Data shown represent
all cell-cell contacts identified in 6 experiments from
2 blood donors. The horizontal line represents the
average of each population. **p < 0.01.

any curative approach. The most widely


accepted mechanism for the generation
of latently infected resting CD4+ T cells is
the return of fully or intermediately acti-
vated cells to a resting state after infection
with HIV-1 (Shan et al., 2017). Based on
B this mechanism, several cell line and
**
60 primary cell in vitro models have been
developed over the years to study HIV-1
transcription and latency (Pace et al.,
Time (min)

40
2011). These models have been instru-
20
mental in the characterization of funda-
mental processes that regulate HIV-1
transcription and latency. However, these
0
No Polar. Polarized models are limited by the use of cells that
may not fully represent the natural pool of
latently infected cells in vivo and may be
2012; Sewald et al., 2012, 2015). Second, the ability of cell-to- biased toward chromatin-dependent repression. Models of
cell transmission to generate infected cells with multiple provi- HIV-1 latency based on direct infection of resting CD4+ T cells
ruses could facilitate recombination, thus increasing viral diver- have been less popular, partly because of the relative resistance
sity (Dang et al., 2004; Del Portillo et al., 2011; Dixit and Perelson, of these cells to HIV-1 infection. These cells have been shown to
2004; Jung et al., 2002). Third, HIV-1 cell-to-cell transmission express some restriction factors that limit HIV infection (Baldauf
could be an important mechanism for the massive depletion of et al., 2012; Zack et al., 2013). Despite the presence of these fac-
CD4+ T cells in untreated infected patients (Doitsh et al., tors, it has been demonstrated that resting cells can still be in-
2010). Our present study proposes that an additional role of fected with HIV directly and at a sufficient frequency to allow
HIV-1 cell-to-cell transmission in vivo is the generation of latently the study of HIV latency in this context (Agosto et al., 2007; Las-
infected cells by direct infection of resting CD4+ T cells. sen et al., 2012; Plesa et al., 2007; Swiggard et al., 2005; Vatakis
Latently infected resting CD4+ T cells represent the most et al., 2007). Studying HIV latency in the context of direct infec-
important source of rebounding virus when treatment is interrup- tion of resting CD4+ T cells can overcome some of the shortcom-
ted (Ruelas and Greene, 2013). The pool of latently infected cells ings of models involving activated T cells and cell lines. In vitro
is comprised of a phenotypically heterogeneous population of models of direct infection of primary resting CD4+ T cells pre-
cells (Churchill et al., 2016). Therefore, a deep understanding serve the natural diversity of cellular phenotypes because artifi-
of how these cells are generated and maintained and how pro- cial manipulations are minimized. Resting CD4+ T cells are
viral latency is regulated is essential for the development of already at a state predisposed to proviral latency; hence,

2094 Cell Reports 24, 2088–2100, August 21, 2018


A HIV Gag-GFP CD4-Alexa647 Merged

P T P T P T

B C D
NS

rCD4T w/ GFP Signal (%)


100
P
T
80

T 60
T
40
T
P
20
0
P Polarized No Polar.

Figure 6. Visualization of Viral Cell-to-Cell Transmission to Resting CD4+ T Cells


(A) Example of polarized HIV-1 assembly as defined by the accumulation of HIV-1 Gag in HIV-1-producing activated cells (P) and CD4 in target cells (T) at the site
of cell-cell contact (arrow).
(B) Example of resting CD4+ T cells carrying particles while in contact with productively infected T cells without polarization of viral assembly.
(C) Example of a resting CD4+ T cell carrying viral particles while in contact with uninfected activated T cells also carrying particles.
(D) The proportion of resting CD4+ T cells that acquired Gag-GFP from polarized viral assembly versus non-polarized viral assembly was quantified by counting
cell-cell contacts.
The scale bars in the images represent 5 mm. Error bars represent the SD of the mean.

studying HIV latency in this context could reveal previously un- ment also help overcome cell resistance to infection and influ-
recognized mechanisms for regulating HIV transcription. The ence the regulation of target cell infection (Shen et al., 2013).
in vitro model presented in this study has the added benefit of Our study explores infection of resting CD4+ cells by cell-to-
incorporating T cell-T cell interactions as part of the process of cell transmission between T cells. These interactions are funda-
generating latent infection. With this model of HIV-1 latency, mentally different than interactions involving dendritic cells or
we demonstrate that productively infected activated CD4+ endothelial cells. T cells typically do not engage in direct antigen
T cells directly transmit HIV-1 to uninfected resting CD4+ presentation with each other; thus, signaling through the T cell
T cells, leading to the generation of pools of latently infected receptors and co-stimulatory receptors is unlikely to be antigen
resting CD4+ T cells. This mechanism for the generation of specific (Len et al., 2017). Therefore, the set of signals involved in
latently infected cells has been underappreciated and likely rep- the process of transmitting HIV-1 to resting cells in the context of
resents an important outcome of HIV-1 spread in vivo before the T cell-T cell interactions is likely unique. It is possible that the
initiation of effective therapy. specific signals and environment created during the transmis-
Initial evidence that cell-contact-mediated transmission of sion of HIV-1 to resting CD4+ T cells from activated T cells will
HIV-1 is relevant for the generation of latently infected cells affect the establishment of inducible latent infection. This lack
was suggested in the context of transmission from dendritic cells of robust T cell signaling, as in the context of antigen presenta-
to resting CD4+ T cells (Evans et al., 2013; Kumar et al., 2015). As tion, likely prevents productive infection and directs proviruses
dendritic cells probe for antigens, they capture and retain full toward a latent state. Our results suggest that proviruses gener-
particles largely in a CD169-dependent fashion (Izquierdo- ated by cell-cell contact between T cells may be harder to induce
Useros et al., 2012; Puryear et al., 2013; Sewald et al., 2015). compared to proviruses generated by cell-free infection. This
As they interact with resting CD4+ T cells during antigen presen- could be a reflection of the route through which latent infection
tation, live particles are transmitted to target cells (McDonald, was generated, the phenotype of cells that harbor latent provi-
2010). In addition, cell-cell contact and the tissue microenviron- ruses, differences in the proportion of defective proviruses, or

Cell Reports 24, 2088–2100, August 21, 2018 2095


A Co-Culture Figure 7. HIV-1 Latent Infection Generated by
Cell-to-Cell Transmission Is Less Inducible

HIV Expression (% Gag+)


Unstimulated Stimulated
3 Than Latent Infection Generated by Cell-
free HIV
(A) Co-cultures of productively infected activated
2
* CD4+ T cells with resting CD4+ T cells were stimulated
FSC-A

with CD3/CD28 beads, IL-2, and IL-7 in the presence


of raltegravir. At 24 hr post-stimulation, HIV-1
1 expression among target cells was assessed by
intracellular HIV-1 Gag staining and flow cytometry
0.651% 1.72% analysis. A representative plot is shown comparing
2 3 4 5 2 3 4 5
0 unstimulated with stimulated cells. A dot plot of the
0 10 10 10 10 0 10 10 10 10 percent of target T cells expressing HIV Gag after

im

28
St

3/
HIV-Gag stimulation or without stimulation is shown. Each

D
ot

+C
symbol represents an independent experiment with

N
Treatment different blood donors. The horizontal line represents
B Cell-Free Spinoculation the average of each population.
(B) Resting CD4+ T cells were spinoculated with
HIV Expression (% Gag+)
Unstimulated Stimulated
15 HIVNL4-3 and incubated for 3 days. At this point, the
** cells were stimulated and analyzed for HIV-1 expres-
sion as in (A). A representative plot is shown
10 comparing unstimulated with stimulated cells.
FSC-A

(C) Resting CD4+ T cells were infected with HIV-1


without spinoculation and incubated for 3 days. At this
1.51% 4.38% 5 point, the cells were stimulated and analyzed for HIV-1
expression as in (A). A representative plot is shown
comparing unstimulated with stimulated cells.
2 3 4 5
0 (D) HIV integration in target resting CD4+ T cells was
2 3 4 5
0 10 10 10 10 0 10 10 10 10 measured under co-culture, spinoculation, and no-
im

28
St

spinoculation infection conditions. Background PCR


3/

HIV-Gag
D
ot

signal was determined based on samples treated


+C
N

Treatment with EFV.


C Cell-Free (E) The proportion of inducible proviruses was calcu-
Unstimulated Stimulated lated from the percent of HIV-positive cells based on
NS
HIV Expression (% Gag+)

flow cytometry after stimulation and after subtracting


3
the background signal from unstimulated cells divided
by the percent HIV-positive cells based on Alu-PCR.
2 This fraction is plotted for 4 independent experiments
FSC-A

0.815% 2.67% done under co-culture conditions and 5 independent


experiments for spinoculation and non-spinoculation
1 infection conditions.
Error bar represent the SD of the mean. In (A), (B), (D),
and (E), *p < 0.05, **p < 0.01.

0 10
2 3
10 10
4
10
5 2
0 10 10
3 4
10
5
10 0
im

28

HIV-Gag
3/
St

D
ot

+C
N

Treatment
D E NS
10 0 0.8
** * *
Ratio Gag+/Provirus

*
(proviruses/cell)
HIV Integration

10 -1 0.6

10 -2 0.4

10 -3 0.2

10 -4 0.0
e

in
C V

el V

e
FV

in
Sp tur

r
re

re
F

F
Sp

Sp
tu
+E

+E
+E

l-F

l-F
ul

ul

F-

el
C
C

in

C
C
o-

o-
C

C
C

Infection Type Infection Type

2096 Cell Reports 24, 2088–2100, August 21, 2018


the presence of multiple proviruses in a small number of in- Microscopy
fected cells (Chavez et al., 2015; Maldarelli, 2016; Rezaei Activated CD4+ T cells were spinoculated with HIVNL4-3-Gag-GFP (Jin et al.,
2009), washed, and incubated for 24 hr at 37 C. Typically, we obtained
et al., 2018; Tsunetsugu-Yokota et al., 2016). Nevertheless,
about 10%–20% GFP-positive cells with our virus stock. Before
our initial observation that latent infection generated by cell- co-culturing with target cells, HIV-infected cells were washed. Infected
to-cell transmission is not easily reversible requires further cells were mixed with resting CD4+ T cells in medium at a 2:1 ratio. Resting
investigation, as current approaches to purging the latent reser- CD4+ T cells were stained with CMAC (Molecular Probes) prior to
voir may not be effective in this context. Our proposed model co-culture. The mixed cells were plated in poly-L-lysine-coated imaging
could be optimized for testing this possibility and could help dishes and incubated at 37 C for 2 hr. Following incubation, the
co-cultured cells were washed and stained for CD4. Finally, the cells
in the search for novel compounds that are more efficient at
were washed, fixed with paraformaldehyde, and imaged by confocal
purging latent provirus.
microscopy.

EXPERIMENTAL PROCEDURES Statistical Analysis


All experiments were repeated at least three times. All data are presented as
Cells mean values ± SD. p values were calculated based on the Mann-Whitney
Peripheral blood mononuclear cells were purified from leukapheresis packs U test or Student’s t test (large sample size) using SPSS software. * denotes
using the Lymphoprep gradient (StemCell Technologies). CD4+ T cells were p < 0.05, ** denotes p < 0.01, and NS denotes non-significant.
negatively selected using the EasySep Human CD4+ T Cell Enrichment Kit
(StemCell Technologies). A fraction of purified CD4+ T cells was stimulated
SUPPLEMENTAL INFORMATION
with phytohaemagglutinin (PHA-P) (Sigma-Aldrich), IL-2, and IL-7 prior to
infection. Another fraction of cells was preserved in medium without exoge-
Supplemental Information includes Supplemental Experimental Procedures,
nous cytokines until needed and then purified the resting cell fraction for co-
one figure, and three videos and can be found with this article online at
culture experiments by depleting cells expressing CD25, CD69, and human
https://doi.org/10.1016/j.celrep.2018.07.079.
leukocyte antigen (HLA)-DR. TZMbl cells were obtained from the NIH AIDS Re-
agents Program.
ACKNOWLEDGMENTS
Viruses
Viruses used for infections of CD4+ T cells were generated by transfection of This work was funded by the amfAR Mathilde Krim Fellowship in Basic
HEK293T cells with plasmids encoding the HIV-1 clones NL4-3 or REJO.c Biomedical Research (award 109263-59-RKRL) and the Immunology Training
(AIDS Reagents Program). Viral supernatants were concentrated by ultra- Grant T32-AI007309. Additional funding was provided by the NIH (awards
centrifugation. Concentrated stocks were subsequently titered in CEM-GFP AI084096, AI097117, and AI118682), the Inflammation Training Grant T32-
cells (AIDS Reagents Program) engineered to express human CCR5. AI89673-5, and the Providence-Boston Center for AIDS Research (CFAR)
(P30-AI042853). We thank Chi Chan and David Levy for advice on transfec-
tions. We thank Nathan Roy, Caitlin Miller, Hisashi Akiyama, and Suryaram
Co-culture Experiments
Gummuluru for advice on imaging cell-cell contacts and reagents. We thank
Activated CD4+ T cells were infected with HIV-1 by spinoculation and incu-
Brian Tilton and the Boston University School of Medicine Flow Cytometry
bated for 48–72 hr at 37 C in the presence of IL-2 and IL-7. Before co-culture,
Core for support with cell sorting and flow cytometry analysis. We thank
infected cells were washed to remove cytokines and unbound viral particles.
Michael Kirber and the Boston University School of Medicine Cellular Imaging
Infected cells were then co-cultured with target resting CD4+ T cells. Parallel
Core for microscopy support.
control co-cultures were treated with EFV (AIDS Reagents Program). The
co-cultures were incubated for 24 hr, treated with SQV (AIDS Reagents Pro-
gram), and incubated for an additional 3 days. At this point, some co-cultures AUTHOR CONTRIBUTIONS
were treated with raltegravir (AIDS Reagents Program) to block further HIV-1
integration and were stimulated with anti-CD3/CD28 beads (Invitrogen) in Conceptualization, L.M.A. and A.J.H.; Investigation, L.M.A. and M.B.H.;
the presence of IL-2 and IL-7 for 24 hr prior to flow cytometry analysis of Writing – Original Draft, L.M.A. and A.J.H.; Supervision, W.M. and A.J.H.;
HIV-1 expression. Other co-cultures were not stimulated but were prepared Funding Acquisition, L.M.A., W.M., and A.J.H.
for sorting of target cells and subsequent PCR analysis.
DECLARATION OF INTERESTS
Cell-free Infection Experiments
Purified resting CD4+ T cells were spinoculated with concentrated HIVNL4-3 or The authors declare no competing interest.
infected without spinoculation. The cells were then washed and resuspended
in medium containing SQV to prevent HIV spread. Control wells were treated Received: January 25, 2018
with EFV. Cells were incubated for 3 days and stimulated with anti-CD3/ Revised: June 1, 2018
CD28 beads (Invitrogen), IL-2, and IL-7 and in the presence of raltegravir for Accepted: July 22, 2018
24 hr prior to flow cytometry analysis of HIV-1 Gag expression. Parallel cultures Published: August 21, 2018
were used for monitoring HIV integration by Alu-PCR.
REFERENCES
Flow Cytometry
Prior to infection by co-culture, target resting CD4+ T cells were stained with Agosto, L.M., Yu, J.J., Dai, J., Kaletsky, R., Monie, D., and O’Doherty, U.
eFluor670 (eBioscience). To monitor HIV-1 Gag expression, co-cultures (2007). HIV-1 integrates into resting CD4+ T cells even at low inoculums as
were fixed with BD CytoFix/CytoPerm buffer (BD Biosciences) and washed demonstrated with an improved assay for HIV-1 integration. Virology 368,
with Perm/Wash buffer (BD Biosciences). Cells were stained with anti-HIV-1 60–72.
Gag antibody in BD Perm/Wash. The cells were then washed with BD Perm/ Agosto, L.M., Yu, J.J., Liszewski, M.K., Baytop, C., Korokhov, N., Humeau,
Wash buffer, resuspended in fluorescence-activated cell sorting (FACS) L.M., and O’Doherty, U. (2009). The CXCR4-tropic human immunodeficiency
buffer, and analyzed by flow cytometry. When monitoring for T cell activation, virus envelope promotes more-efficient gene delivery to resting CD4+ T cells
the cells were stained for CD25, CD69, and HLA-DR expression. Viability was than the vesicular stomatitis virus glycoprotein G envelope. J. Virol. 83,
monitored by staining cells with the Zombie Fixable Viability Kit (BioLegend). 8153–8162.

Cell Reports 24, 2088–2100, August 21, 2018 2097


Agosto, L.M., Zhong, P., Munro, J., and Mothes, W. (2014). Highly active anti- Dixit, N.M., and Perelson, A.S. (2004). Multiplicity of human immunodeficiency
retroviral therapies are effective against HIV-1 cell-to-cell transmission. PLoS virus infections in lymphoid tissue. J. Virol. 78, 8942–8945.
Pathog. 10, e1003982. Doitsh, G., Cavrois, M., Lassen, K.G., Zepeda, O., Yang, Z., Santiago, M.L.,
Agosto, L.M., Gagne, M., and Henderson, A.J. (2015a). Impact of chromatin on Hebbeler, A.M., and Greene, W.C. (2010). Abortive HIV infection mediates
HIV replication. Genes (Basel) 6, 957–976. CD4 T cell depletion and inflammation in human lymphoid tissue. Cell 143,
Agosto, L.M., Uchil, P.D., and Mothes, W. (2015b). HIV cell-to-cell transmis- 789–801.
sion: effects on pathogenesis and antiretroviral therapy. Trends Microbiol. Douek, D.C., Brenchley, J.M., Betts, M.R., Ambrozak, D.R., Hill, B.J., Oka-
23, 289–295. moto, Y., Casazza, J.P., Kuruppu, J., Kunstman, K., Wolinsky, S., et al.
Baldauf, H.M., Pan, X., Erikson, E., Schmidt, S., Daddacha, W., Burggraf, M., (2002). HIV preferentially infects HIV-specific CD4+ T cells. Nature 417, 95–98.
Schenkova, K., Ambiel, I., Wabnitz, G., Gramberg, T., et al. (2012). SAMHD1 Duncan, C.J., Williams, J.P., Schiffner, T., Gärtner, K., Ochsenbauer, C.,
restricts HIV-1 infection in resting CD4(+) T cells. Nat. Med. 18, 1682–1687. Kappes, J., Russell, R.A., Frater, J., and Sattentau, Q.J. (2014). High-multiplic-
Brandenberg, O.F., Rusert, P., Magnus, C., Weber, J., Boni, J., Gunthard, H.F., ity HIV-1 infection and neutralizing antibody evasion mediated by the macro-
Regoes, R.R., and Trkola, A. (2014). Partial rescue of V1V2 mutant infectivity by phage-T cell virological synapse. J. Virol. 88, 2025–2034.
HIV-1 cell-cell transmission supports the domain’s exceptional capacity for
Eckstein, D.A., Penn, M.L., Korin, Y.D., Scripture-Adams, D.D., Zack, J.A.,
sequence variation. Retrovirology 11, 75.
Kreisberg, J.F., Roederer, M., Sherman, M.P., Chin, P.S., and Goldsmith,
Brenchley, J.M., Hill, B.J., Ambrozak, D.R., Price, D.A., Guenaga, F.J., Ca- M.A. (2001). HIV-1 actively replicates in naive CD4(+) T cells residing within hu-
sazza, J.P., Kuruppu, J., Yazdani, J., Migueles, S.A., Connors, M., et al. man lymphoid tissues. Immunity 15, 671–682.
(2004). T-cell subsets that harbor human immunodeficiency virus (HIV) in vivo:
Evans, V.A., Kumar, N., Filali, A., Procopio, F.A., Yegorov, O., Goulet, J.P.,
implications for HIV pathogenesis. J. Virol. 78, 1160–1168.
Saleh, S., Haddad, E.K., da Fonseca Pereira, C., Ellenberg, P.C., et al.
Bukrinsky, M.I., Stanwick, T.L., Dempsey, M.P., and Stevenson, M. (1991). (2013). Myeloid dendritic cells induce HIV-1 latency in non-proliferating
Quiescent T lymphocytes as an inducible virus reservoir in HIV-1 infection. Sci- CD4+ T cells. PLoS Pathog. 9, e1003799.
ence 254, 423–427.
Finzi, D., Hermankova, M., Pierson, T., Carruth, L.M., Buck, C., Chaisson, R.E.,
Chavez, L., Calvanese, V., and Verdin, E. (2015). HIV latency is established
Quinn, T.C., Chadwick, K., Margolick, J., Brookmeyer, R., et al. (1997). Identi-
directly and early in both resting and activated primary CD4 T cells. PLoS
fication of a reservoir for HIV-1 in patients on highly active antiretroviral ther-
Pathog. 11, e1004955.
apy. Science 278, 1295–1300.
Chen, P., Hu €bner, W., Spinelli, M.A., and Chen, B.K. (2007). Predominant
Gupta, P., Balachandran, R., Ho, M., Enrico, A., and Rinaldo, C. (1989). Cell-to-
mode of human immunodeficiency virus transfer between T cells is mediated
cell transmission of human immunodeficiency virus type 1 in the presence of
by sustained Env-dependent neutralization-resistant virological synapses.
azidothymidine and neutralizing antibody. J. Virol. 63, 2361–2365.
J. Virol. 81, 12582–12595.
Chomont, N., El-Far, M., Ancuta, P., Trautmann, L., Procopio, F.A., Yassine- Izquierdo-Useros, N., Lorizate, M., Puertas, M.C., Rodriguez-Plata, M.T.,
Diab, B., Boucher, G., Boulassel, M.R., Ghattas, G., Brenchley, J.M., et al. Zangger, N., Erikson, E., Pino, M., Erkizia, I., Glass, B., Clotet, B., et al.
(2009). HIV reservoir size and persistence are driven by T cell survival and ho- (2012). Siglec-1 is a novel dendritic cell receptor that mediates HIV-1 trans-
meostatic proliferation. Nat. Med. 15, 893–900. infection through recognition of viral membrane gangliosides. PLoS Biol. 10,
e1001448.
Chun, T.W., Carruth, L., Finzi, D., Shen, X., DiGiuseppe, J.A., Taylor, H., Her-
mankova, M., Chadwick, K., Margolick, J., Quinn, T.C., et al. (1997a). Quanti- Jin, J., Sturgeon, T., Weisz, O.A., Mothes, W., and Montelaro, R.C. (2009).
fication of latent tissue reservoirs and total body viral load in HIV-1 infection. HIV-1 matrix dependent membrane targeting is regulated by Gag mRNA traf-
Nature 387, 183–188. ficking. PLoS ONE 4, e6551.
Chun, T.W., Stuyver, L., Mizell, S.B., Ehler, L.A., Mican, J.A., Baseler, M., Johnson, D.C., and Huber, M.T. (2002). Directed egress of animal viruses pro-
Lloyd, A.L., Nowak, M.A., and Fauci, A.S. (1997b). Presence of an inducible motes cell-to-cell spread. J. Virol. 76, 1–8.
HIV-1 latent reservoir during highly active antiretroviral therapy. Proc. Natl. Jolly, C., Kashefi, K., Hollinshead, M., and Sattentau, Q.J. (2004). HIV-1 cell to
Acad. Sci. USA 94, 13193–13197. cell transfer across an Env-induced, actin-dependent synapse. J. Exp. Med.
Chun, T.W., Engel, D., Berrey, M.M., Shea, T., Corey, L., and Fauci, A.S. (1998). 199, 283–293.
Early establishment of a pool of latently infected, resting CD4(+) T cells during
Jolly, C., Booth, N.J., and Neil, S.J. (2010). Cell-cell spread of human immuno-
primary HIV-1 infection. Proc. Natl. Acad. Sci. USA 95, 8869–8873.
deficiency virus type 1 overcomes tetherin/BST-2-mediated restriction in
Churchill, M.J., Deeks, S.G., Margolis, D.M., Siliciano, R.F., and Swanstrom, T cells. J. Virol. 84, 12185–12199.
R. (2016). HIV reservoirs: what, where and how to target them. Nat. Rev. Micro-
Jung, A., Maier, R., Vartanian, J.P., Bocharov, G., Jung, V., Fischer, U., Meese,
biol. 14, 55–60.
E., Wain-Hobson, S., and Meyerhans, A. (2002). Recombination: Multiply in-
Dai, J., Agosto, L.M., Baytop, C., Yu, J.J., Pace, M.J., Liszewski, M.K., and fected spleen cells in HIV patients. Nature 418, 144.
O’Doherty, U. (2009). Human immunodeficiency virus integrates directly into
naive resting CD4+ T cells but enters naive cells less efficiently than memory Kinter, A., Moorthy, A., Jackson, R., and Fauci, A.S. (2003). Productive HIV
cells. J. Virol. 83, 4528–4537. infection of resting CD4+ T cells: role of lymphoid tissue microenvironment
and effect of immunomodulating agents. AIDS Res. Hum. Retroviruses 19,
Dang, Q., Chen, J., Unutmaz, D., Coffin, J.M., Pathak, V.K., Powell, D., Kewal-
847–856.
Ramani, V.N., Maldarelli, F., and Hu, W.S. (2004). Nonrandom HIV-1 infection
and double infection via direct and cell-mediated pathways. Proc. Natl. Acad. Korin, Y.D., and Zack, J.A. (1998). Progression to the G1b phase of the cell cy-
Sci. USA 101, 632–637. cle is required for completion of human immunodeficiency virus type 1 reverse
transcription in T cells. J. Virol. 72, 3161–3168.
Davey, R.T., Jr., Bhat, N., Yoder, C., Chun, T.-W., Metcalf, J.A., Dewar, R., Na-
tarajan, V., Lempicki, R.A., Adelsberger, J.W., Miller, K.D., et al. (1999). HIV-1 Kumar, N.A., Cheong, K., Powell, D.R., da Fonseca Pereira, C., Anderson, J.,
and T cell dynamics after interruption of highly active antiretroviral therapy Evans, V.A., Lewin, S.R., and Cameron, P.U. (2015). The role of antigen pre-
(HAART) in patients with a history of sustained viral suppression. Proc. Natl. senting cells in the induction of HIV-1 latency in resting CD4(+) T-cells. Retro-
Acad. Sci. USA 96, 15109–15114. virology 12, 76.
Del Portillo, A., Tripodi, J., Najfeld, V., Wodarz, D., Levy, D.N., and Chen, B.K. Lassen, K.G., Hebbeler, A.M., Bhattacharyya, D., Lobritz, M.A., and Greene,
(2011). Multiploid inheritance of HIV-1 during cell-to-cell infection. J. Virol. 85, W.C. (2012). A flexible model of HIV-1 latency permitting evaluation of many
7169–7176. primary CD4 T-cell reservoirs. PLoS ONE 7, e30176.

2098 Cell Reports 24, 2088–2100, August 21, 2018


Law, K.M., Komarova, N.L., Yewdall, A.W., Lee, R.K., Herrera, O.L., Wodarz, use CD169-mediated trans-infection of permissive lymphocytes to establish
D., and Chen, B.K. (2016). In vivo HIV-1 cell-to-cell transmission promotes infection. Science 350, 563–567.
multicopy micro-compartmentalized infection. Cell Rep. 15, 2771–2783. Shan, L., Deng, K., Gao, H., Xing, S., Capoferri, A.A., Durand, C.M., Rabi, S.A.,
Len, A.C.L., Starling, S., Shivkumar, M., and Jolly, C. (2017). HIV-1 activates Laird, G.M., Kim, M., Hosmane, N.N., et al. (2017). Transcriptional reprogram-
T cell signaling independently of antigen to drive viral spread. Cell Rep. 18, ming during effector-to-memory transition renders CD4+ T cells permissive for
1062–1074. latent HIV-1 infection. Immunity 47, 766–775.e3.
Liszewski, M.K., Yu, J.J., and O’Doherty, U. (2009). Detecting HIV-1 integration Shen, A., Baker, J.J., Scott, G.L., Davis, Y.P., Ho, Y.Y., and Siliciano, R.F.
by repetitive-sampling Alu-gag PCR. Methods 47, 254–260. (2013). Endothelial cell stimulation overcomes restriction and promotes pro-
Maldarelli, F. (2016). The role of HIV integration in viral persistence: no more ductive and latent HIV-1 infection of resting CD4+ T cells. J. Virol. 87, 9768–
whistling past the proviral graveyard. J. Clin. Invest. 126, 438–447. 9779.

Maldarelli, F., Wu, X., Su, L., Simonetti, F.R., Shao, W., Hill, S., Spindler, J., Fer- Sherer, N.M., Jin, J., and Mothes, W. (2010). Directional spread of surface-
ris, A.L., Mellors, J.W., Kearney, M.F., et al. (2014). HIV latency. Specific HIV associated retroviruses regulated by differential virus-cell interactions.
integration sites are linked to clonal expansion and persistence of infected J. Virol. 84, 3248–3258.
cells. Science 345, 179–183. Sigal, A., Kim, J.T., Balazs, A.B., Dekel, E., Mayo, A., Milo, R., and Baltimore, D.
Mathez, D., Schinazi, R.F., Liotta, D.C., and Leibowitch, J. (1993). Infectious (2011). Cell-to-cell spread of HIV permits ongoing replication despite antiretro-
amplification of wild-type human immunodeficiency virus from patients’ lym- viral therapy. Nature 477, 95–98.
phocytes and modulation by reverse transcriptase inhibitors in vitro. Antimi- Sourisseau, M., Sol-Foulon, N., Porrot, F., Blanchet, F., and Schwartz, O.
crob. Agents Chemother. 37, 2206–2211. (2007). Inefficient human immunodeficiency virus replication in mobile lympho-
Mbonye, U., and Karn, J. (2017). The molecular basis for human immunodefi- cytes. J. Virol. 81, 1000–1012.
ciency virus latency. Annu. Rev. Virol. 4, 261–285. Stevenson, M., Stanwick, T.L., Dempsey, M.P., and Lamonica, C.A. (1990).
McDonald, D. (2010). Dendritic cells and HIV-1 trans-infection. Viruses 2, HIV-1 replication is controlled at the level of T cell activation and proviral inte-
1704–1717. gration. EMBO J. 9, 1551–1560.
Strain, M.C., Gu€nthard, H.F., Havlir, D.V., Ignacio, C.C., Smith, D.M., Leigh-
Murooka, T.T., Deruaz, M., Marangoni, F., Vrbanac, V.D., Seung, E., von An-
drian, U.H., Tager, A.M., Luster, A.D., and Mempel, T.R. (2012). HIV-infected Brown, A.J., Macaranas, T.R., Lam, R.Y., Daly, O.A., Fischer, M., et al.
T cells are migratory vehicles for viral dissemination. Nature 490, 283–287. (2003). Heterogeneous clearance rates of long-lived lymphocytes infected
with HIV: intrinsic stability predicts lifelong persistence. Proc. Natl. Acad.
Nishimura, Y., Brown, C.R., Mattapallil, J.J., Igarashi, T., Buckler-White, A., La- Sci. USA 100, 4819–4824.
font, B.A., Hirsch, V.M., Roederer, M., and Martin, M.A. (2005). Resting naive
CD4+ T cells are massively infected and eliminated by X4-tropic simian-human Sun, Y., and Clark, E.A. (1999). Expression of the c-myc proto-oncogene is
immunodeficiency viruses in macaques. Proc. Natl. Acad. Sci. USA 102, 8000– essential for HIV-1 infection in activated T cells. J. Exp. Med. 189, 1391–1398.
8005. Swiggard, W.J., Baytop, C., Yu, J.J., Dai, J., Li, C., Schretzenmair, R., Theodo-
Ostrowski, M.A., Chun, T.-W., Justement, S.J., Motola, I., Spinelli, M.A., Adels- sopoulos, T., and O’Doherty, U. (2005). Human immunodeficiency virus type 1
berger, J., Ehler, L.A., Mizell, S.B., Hallahan, C.W., and Fauci, A.S. (1999). Both can establish latent infection in resting CD4+ T cells in the absence of acti-
memory and CD45RA+/CD62L+ naive CD4(+) T cells are infected in human im- vating stimuli. J. Virol. 79, 14179–14188.
munodeficiency virus type 1-infected individuals. J. Virol. 73, 6430–6435. Tabler, C.O., Lucera, M.B., Haqqani, A.A., McDonald, D.J., Migueles, S.A.,
Pace, M.J., Agosto, L., Graf, E.H., and O’Doherty, U. (2011). HIV reservoirs and Connors, M., and Tilton, J.C. (2014). CD4+ memory stem cells are infected
latency models. Virology 411, 344–354. by HIV-1 in a manner regulated in part by SAMHD1 expression. J. Virol. 88,
4976–4986.
Phillips, D.M. (1994). The role of cell-to-cell transmission in HIV infection. AIDS
8, 719–731. Titanji, B.K., Aasa-Chapman, M., Pillay, D., and Jolly, C. (2013). Protease inhib-
itors effectively block cell-to-cell spread of HIV-1 between T cells. Retrovirol-
Plesa, G., Dai, J., Baytop, C., Riley, J.L., June, C.H., and O’Doherty, U. (2007). ogy 10, 161.
Addition of deoxynucleosides enhances human immunodeficiency virus type 1
integration and 2LTR formation in resting CD4+ T cells. J. Virol. 81, 13938– Tsunetsugu-Yokota, Y., Kobayahi-Ishihara, M., Wada, Y., Terahara, K., Take-
13942. yama, H., Kawana-Tachikawa, A., Tokunaga, K., Yamagishi, M., Martinez,
J.P., and Meyerhans, A. (2016). Homeostatically maintained resting naive
Puryear, W.B., Akiyama, H., Geer, S.D., Ramirez, N.P., Yu, X., Reinhard, B.M., CD4+ T cells resist latent HIV reactivation. Front. Microbiol. 7, 1944.
and Gummuluru, S. (2013). Interferon-inducible mechanism of dendritic cell-
mediated HIV-1 dissemination is dependent on Siglec-1/CD169. PLoS Unutmaz, D., KewalRamani, V.N., Marmon, S., and Littman, D.R. (1999). Cyto-
Pathog. 9, e1003291. kine signals are sufficient for HIV-1 infection of resting human T lymphocytes.
J. Exp. Med. 189, 1735–1746.
Rezaei, S.D., Lu, H.K., Chang, J.J., Rhodes, A., Lewin, S.R., and Cameron,
Vatakis, D.N., Bristol, G., Wilkinson, T.A., Chow, S.A., and Zack, J.A. (2007).
P.U. (2018). The pathway to establishing HIV latency is critical to how latency
Immediate activation fails to rescue efficient human immunodeficiency virus
is maintained and reversed. J. Virol. 92, e02225-17.
replication in quiescent CD4+ T cells. J. Virol. 81, 3574–3582.
Ruelas, D.S., and Greene, W.C. (2013). An integrated overview of HIV-1 la-
Wagner, T.A., McLaughlin, S., Garg, K., Cheung, C.Y., Larsen, B.B., Styrchak,
tency. Cell 155, 519–529.
S., Huang, H.C., Edlefsen, P.T., Mullins, J.I., and Frenkel, L.M. (2014). HIV la-
Saleh, S., Solomon, A., Wightman, F., Xhilaga, M., Cameron, P.U., and Lewin, tency. Proliferation of cells with HIV integrated into cancer genes contributes
S.R. (2007). CCR7 ligands CCL19 and CCL21 increase permissiveness of to persistent infection. Science 345, 570–573.
resting memory CD4+ T cells to HIV-1 infection: a novel model of HIV-1 la-
Whitney, J.B., Hill, A.L., Sanisetty, S., Penaloza-MacMaster, P., Liu, J., Shetty,
tency. Blood 110, 4161–4164.
M., Parenteau, L., Cabral, C., Shields, J., Blackmore, S., et al. (2014). Rapid
Sattentau, Q. (2008). Avoiding the void: cell-to-cell spread of human viruses. seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature
Nat. Rev. Microbiol. 6, 815–826. 512, 74–77.
Sewald, X., Gonzalez, D.G., Haberman, A.M., and Mothes, W. (2012). In vivo €nthard, H.F., Havlir, D.V., Ignacio, C.C., Spina,
Wong, J.K., Hezareh, M., Gu
imaging of virological synapses. Nat. Commun. 3, 1320. C.A., and Richman, D.D. (1997). Recovery of replication-competent HIV
Sewald, X., Ladinsky, M.S., Uchil, P.D., Beloor, J., Pi, R., Herrmann, C., Mota- despite prolonged suppression of plasma viremia. Science 278, 1291–
medi, N., Murooka, T.T., Brehm, M.A., Greiner, D.L., et al. (2015). Retroviruses 1295.

Cell Reports 24, 2088–2100, August 21, 2018 2099


Zack, J.A., Haislip, A.M., Krogstad, P., and Chen, I.S.Y. (1992). Incompletely Zhang, Z., Schuler, T., Zupancic, M., Wietgrefe, S., Staskus, K.A., Reimann,
reverse-transcribed human immunodeficiency virus type 1 genomes in quies- K.A., Reinhart, T.A., Rogan, M., Cavert, W., Miller, C.J., et al. (1999). Sexual
cent cells can function as intermediates in the retroviral life cycle. J. Virol. 66, transmission and propagation of SIV and HIV in resting and activated CD4+
1717–1725. T cells. Science 286, 1353–1357.
Zack, J.A., Kim, S.G., and Vatakis, D.N. (2013). HIV restriction in quiescent Zhong, P., Agosto, L.M., Ilinskaya, A., Dorjbal, B., Truong, R., Derse, D., Uchil,
CD4+ T cells. Retrovirology 10, 37. P.D., Heidecker, G., and Mothes, W. (2013a). Cell-to-cell transmission can
Zerbato, J.M., Serrao, E., Lenzi, G., Kim, B., Ambrose, Z., Watkins, S.C., En- overcome multiple donor and target cell barriers imposed on cell-free HIV.
gelman, A.N., and Sluis-Cremer, N. (2016). Establishment and reversal of PLoS ONE 8, e53138.
HIV-1 latency in naive and central memory CD4+ T cells in vitro. J. Virol. 90, Zhong, P., Agosto, L.M., Munro, J.B., and Mothes, W. (2013b). Cell-to-cell
8059–8073. transmission of viruses. Curr. Opin. Virol. 3, 44–50.

2100 Cell Reports 24, 2088–2100, August 21, 2018

You might also like