You are on page 1of 7

Special Issue: Microbial Endurance

Review
TFH in HIV Latency and as
Sources of Replication-
Competent Virus
Brodie Miles1 and Elizabeth Connick1,*
During untreated disease, HIV replication is concentrated within T follicular
Trends
helper cells (TFH). Heightened permissiveness, the presence of highly infectious
TFH are the major HIV-producing cells
virions on follicular dendritic cells (FDCs), low frequencies of virus-specic in untreated disease.
cytotoxic T lymphocytes (CTLs) in B cell follicles, expansions in TFH, and TFH
Heightened TFH permissivity, virion
dysfunction, all likely promote replication in TFH. Limited data suggest that retention by FDCs, exclusion of CTLs
memory TFH play a role in the latent or subclinical reservoir of HIV during from the follicle, expansion of the TFH
antiretroviral therapy (ART), potentially for many of the same reasons. A better subset, and TFH dysfunction, all con-
tribute to HIV replication in TFH.
understanding of the role of memory TFH and FDC-bound virions in promoting
recrudescent viremia in the setting of ART cessation is essential. Studies that Limited data suggest that follicular
reservoirs of HIV, including latently
target follicular virus reservoirs are needed to determine their role in HIV latency
infected memory TFH and FDC-bound
and to suggest successful cure strategies. virions, exist in treated disease.

Strategies that target follicular reser-


voirs of HIV could be used to dissect
Role of TFH in HIV Replication in Untreated Disease
out their role in the latent reservoir and
TFH are a specialized subset of CD4+ helper T cells that express CXCR5, migrate into B cell HIV persistence.
follicles, and promote B cell maturation and antibody production during infections (reviewed in
[1]). In untreated, asymptomatic HIV infection, TFH serve as the major site of HIV infection and
replication (Figure 1) [25]. A median of 6075% of HIV-producing cells are located within B cell
follicles in lymph nodes from untreated, asymptomatically infected individuals [2,4], and a CD4+
T cell located within a B cell follicle is 40 times more likely to be productively infected than a
CD4+ T cell located outside of the follicle [4]. Similarly, in chronically SIV-infected rhesus
macaques without simian AIDS (SAIDS), the majority of SIV-producing cells are located within
follicles [6,7]. Even after normalizing for differences in memory cells between the follicular and
extrafollicular compartments, follicular CD4+ T cells are a median of 6.5 times more likely to
be producing SIV RNA than are extrafollicular CD4+ T cells [6]. Interestingly, in nonpathogenic
SIV infection in sooty mangabeys, a follicular concentration of virus replication is not observed
[7], suggesting that productive TFH infection may be a critical driver of HIV and SIV
immunopathogenesis.

Mechanisms that promote HIV replication within TFH are not fully understood. TFH from human
tonsils are highly permissive to both CCR5- and CXCR4-tropic HIV compared to other CD4+
T cell subsets ex vivo [8,9]. The heightened permissivity of tonsillar TFH cannot be fully explained 1
Division of Infectious Diseases,
by differences in memory subsets, cellular activation, or chemokine coreceptor expression [9]. University of Colorado Denver, Aurora
Importantly, in an HIV model system using humanized mice, TFH rapidly accumulate in gut and CO 80045, USA

female reproductive tract mucosal tissues and are the most permissive CD4+ T cell subset to HIV
[10], suggesting that gut and vaginal TFH may play a key role in establishment of HIV infection as *Correspondence:
well as ongoing virus replication. Liz.Connick@denver.edu (E. Connick).

338 Trends in Microbiology, May 2016, Vol. 24, No. 5 http://dx.doi.org/10.1016/j.tim.2016.02.006


2016 Elsevier Ltd. All rights reserved.
Chronically HIV-infected 1 4
HIV+ HIV+
lymph node
CTL T
T Cell T

T HIV+ T


T

HIV+ Uninfected and HIV-producing


CTL T Cell
T accumulate

HIV-specic CTL kill


virus-producing T cells

2 3
FDC
FDC

CTL
HIV+ HIV+
T
Follicle T T
Extrafollicular
zone CTL fail to migrate
into the follicle Angen exposure maintains T phenotype
and T acquire infecous HIV from FDC

Extrafollicular zone Follicle

Figure 1. Model of T Follicular Helper Cells (TFH) Accumulation in Chronic, Untreated HIV Infection. HIV-specic
cytotoxic T lymphocytes (CTLs) recognize and kill virus-producing T cells (HIV+ T cell) in the extrafollicular zone (1), but are
found in low numbers within the follicle due to low CXCR5 expression (2). Within the follicle, TFH receive both activation
signals and infectious HIV from interactions with follicular dendritic cells (FDCs) (3). TFH, including HIV-producing TFH (HIV+),
accumulate within the follicle (4).

The presence of an FDC network and large amounts of FDC-associated HIV virions adjacent to
TFH within germinal centers (GC) [1113] is one contributing factor predisposing TFH to high
levels of HIV infection and replication. This extracellular burden of virions is bound to FDCs via
antibody through complement and FC receptors [14] and appears shortly after infection [15]. In
chronic disease, the amount of viral RNA (vRNA) associated with FDC is 10- to 40-fold more than
is found in lymphoid mononuclear cells [12]. FDC harbor archived virus from the host [16], and
these virions are potently infectious to CD4+ T cells, even in the presence of neutralizing
antibodies [13]. FDCs further promote HIV replication within TFH by upregulating virus transcrip-
tion through release of tumor necrosis factor-/ (TNF-/) [8].

High concentrations of virus replication within TFH are further promoted by a paucity of virus-
specic CTLs within B cell follicles in both HIV [4] and SIV infection [6,17]. Few SIV-specic CTLs
express the follicular homing molecule CXCR5 in the absence of the extrafollicular retention
molecule CCR7, which restricts most from entering the follicle [6]. SIV replication is widespread
throughout lymphoid tissues, and viral replication is not concentrated in the follicle during acute
infection, prior to when the CTL response has evolved to control the virus [6]. Similarly,
compartmentalization of virus replication in TFH is attenuated during AIDS, when the virus-
specic CTL response is known to wane [6], and ablated following CD8 T cell depletion in rhesus
macaques [18]. Thus, other lymphoid cells besides TFH are capable of replicating HIV-1, but
largely restricted from doing so during asymptomatic disease by the presence of CTLs.

Remarkably, despite the fact that TFH are the major virus-producing cells in asymptomatic
disease, they increase in number during early and mid-stages of chronic HIV [3,19] and SIV

Trends in Microbiology, May 2016, Vol. 24, No. 5 339


infection [20]. It was shown in rhesus macaques that acute SIV infection results in the rapid
formation of GC and an accumulation of TFH coinciding with high levels of p27 expression in the
follicle [21]. Furthermore, in lymph nodes from chronically HIV-infected individuals, virus-specic
TFH, particularly HIV Gag-specic TFH, are expanded [19]. Thus, at least part of TFH expansion is
due to HIV antigen, as CD4+ T cells require antigen exposure for most cell division [22] and
antigen persistence in the GC is required to sustain TFH phenotypes [23]. Within the GC, TFH are
chronically exposed to HIV virions and antigens through interactions with B cells [24] and FDCs
[11,12]. Circulating TFH are decreased in untreated chronically-HIV infected individuals [25],
suggesting that the GC offers a unique environment allowing for TFH expansion. It should be
noted that, in advanced infection, TFH expansions and GC morphology are lost, and this is
associated with the onset of SAIDS in rhesus macaques [26] and AIDS in humans [27].

Non-antigen-specic inuences may promote expansion and persistence of TFH in HIV and SIV
infection in early stages of disease as well. Alterations of cytokine production observed during
HIV and SIV infection, such as decreased interleukin-2 (IL-2) [28] and increased IL-6 [20,29] and
interferon-g (IFN-g) [30], promote TFH expansions [1]. T follicular regulatory cells (TFR) are a subset
of follicular cells that directly regulate numbers and function of TFH during HIV and SIV infection
[3134]. Two studies linked relative decreases of TFR to expansions of TFH in the SIV-infected
rhesus macaque model [31,32]. Increased cell survival may contribute to the persistence of TFH
as well. CXCR5+ cells in healthy human tonsils express elevated levels of the antiapoptotic
protein Bcl-2, and in the context of ex vivo R5 infection, Bcl-2 is upregulated in productively
infected cells [35]. Interestingly, the depletion of CD8 T cells in untreated SIV infection leads to
similar levels of replication in lymphoid memory TFH and memory non-TFH; however, the memory
TFH contain higher levels of RNA and DNA copies up to 135 days after depletion and after CD8
T cell recovery [18], suggesting possible preferential survival of TFH.

HIV infection not only leads to alterations in TFH numbers, but also changes in TFH function. TFH
from lymph nodes of untreated individuals demonstrate expansions of HIV-specic IL-21+,
INF-g+, and TNF-/+ TFH compared to treated individuals [19]. Furthermore, B cells from
untreated individuals demonstrate a skewed phenotype [19]. Hypergammaglobulinemia is a
well described phenomenon in untreated HIV-infected individuals, and Bcl6 expression in TFH
correlates with immunoglobulin G (IgG) levels [19]. Nevertheless, TFH from untreated HIV patients
are unable to stimulate robust IgG production in vitro [36], suggesting signicant impairments in
TFH in the context of untreated infection, which is consistent with known clinical impairments in
responses to vaccines. In vitro, impairments in the ability of TFH to stimulate IgG are reversed by
supplementation with IL-21 or blockade of PD-1 ligation on TFH [36]. Circulating IL-21+ CD4+
T cells were shown to be transcriptionally equivalent to TFH in seronegative individuals and
peripheral HIV-specic IL-21+ TFH in untreated HIV+ subjects were able to stimulate autologous
CD8+ T cell activation and B cell class switching [37]. Expansions in TFR occur in the context of
both HIV and SIV infection, and these cells impair the ability of TFH to secrete IL-21 and IL-4 [33],
suggesting another potential mechanism for TFH dysfunction.

Follicular HIV Reservoirs during Treated Disease


A stable and inducible latent viral reservoir within memory CD4+ T cells is maintained in HIV-
infected individuals despite years of ART that renders plasma viremia undetectable [3840].
Furthermore, low frequencies of vRNA+ lymphocytes persist in peripheral blood [41] and lymph
nodes of treated individuals [42] undergoing successful long-term ART. Virus replication
rebounds from multiple foci in secondary lymphoid tissues once ART is stopped in both
HIV-infected humans and SIV-infected rhesus macaques [43,44]. Characteristics of the viral
reservoir that is the source of recrudescent viremia are incompletely understood, and this is a
major barrier to the development of an HIV cure. Furthermore, whether virus replication is
ongoing in the context of ART is a matter of controversy. The presence of vRNA+ cells during

340 Trends in Microbiology, May 2016, Vol. 24, No. 5


ART could be due to reactivation of latently infected cells or, alternatively, ongoing rounds of virus
replication. Although drug concentrations have been reported to be suboptimal in the lymph
node [45], curiously little evidence for virus evolution has been found in most studies in patients
whose viral loads remained well suppressed [4648]. Furthermore, ART-related resistance
mutations do not develop after long-term virologic suppression [47,49], which one would expect
to occur if indeed drug concentrations in lymph nodes were suboptimal. Indeed, founder
variants are the main HIV strains that emerge from lymphatic tissues during treatment interrup-
tion [44], and increasing data suggest that homeostatic proliferation of viral DNA+ (vDNA) cells is
the major factor underlying persistent viral DNA levels [48,50].

The fact that TFH display antiapoptotic properties, as discussed above, despite being the major
virus-producing cell subset in untreated, asymptomatic HIV, suggests that they could harbor a
signicant amount of the latent viral DNA during treated disease. Indeed, in ART-suppressed
SIV-infected rhesus macaques, the major vRNA+ cells in lymphoid tissues are TFH, as deter-
mined by cell sorting studies [18], although vDNA was not concentrated in this subset. Virus
replication in these cells could represent reactivation of latently infected memory TFH. The
existence of memory TFH in humans has only recently been recognized [1,51], and they have
not been extensively studied in the context of HIV or SIV infection. TFH transcription factors may
promote establishment of HIV latency. The HIV long terminal repeat (LTR) contains binding sites
for the master TFH transcription factor Bcl6 that allows Bcl6 to repress HIV transcription [52], and
it has been speculated that Bcl6 supports HIV latency [53]. Memory TFH can be long-lived and
primarily have a central memory phenotype, similar to the resting CD4+ T cell reservoir that
harbors the majority of HIV DNA in treated individuals [50]. One recent study reported that the
majority of HIV-1 DNA in peripheral blood was harbored by central memory TFH in subjects with
sustained virologic suppression on ART [54]. Nevertheless, the precise phenotype of peripheral
TFH is somewhat controversial [55] and until an accurate phenotype is determined, identifying the
role of TFH in the latent HIV reservoir using peripheral blood will be challenging.

During ART, TFH numbers decrease relative to untreated disease, but remain elevated compared
to healthy individuals [19]. Phenotypically, lymph node TFH from treated HIV+ subjects have lower
activation rates as evidenced by decreased Bcl6 expression and fewer HIV-specic IL-21+ TFH,
INF-g+ TFH, and TNF-/+ TFH [19]. The expansion of TFH in both HIV+ untreated and treated
subjects, compared to healthy controls, is also associated with an increased transitional B cell
phenotype and lower memory B cell formation [19]. The number of functional circulating TFH is
decreased in untreated HIV+ subjects, and TFH numbers and function slightly recover in treated
HIV+ subjects [25]. However, TFH function is still incomplete, and HIV+ subjects still have
functional impairments [19] and poor vaccine responses [56,57]. TFH function has also been
found to be crucial for subsequent vaccine responses in treated HIV+ subjects, as responses to
u vaccination were directly dependent on the ability of TFH to proliferate, express inducible T cell
co-stimulator (ICOS), and produce IL-21 [58].

An alternative or additional explanation for the presence of HIV RNA+ cells in TFH in the context of
suppressive ART is that they represent new infection from virus archived on FDCs. Following
6 months of ART, FDC-bound virions decrease but are nonetheless detectable; the residual
number of copies of HIV RNA present on FDCs was estimated to be nearly 108 virions in an
average individual [59]. How long FDC-bound virions remain infectious is unknown. In a
nonpermissive mouse model, FDC-bound HIV remained infectious for the maximum period
studied, which was 9 months [60]. Although antiretroviral drugs that block virus life cycle at the
stages of entry, reverse transcription, and integration prevent most infections, these may not be
fully effective, resulting in occasional virus integration and replication. Thus, FDC-bound HIV
could potentially provide a low level of new infections that manifest as founder virus, due to the
archived nature of FDC-bound virions [16].

Trends in Microbiology, May 2016, Vol. 24, No. 5 341


Eliminating Follicular HIV Reservoirs
To date only one person has been successfully cured of HIV infection after receiving a bone marrow
transplant with donor cells that were genetically resistant to HIV infection [61]. Multiple other
attempts to achieve a cure or reduce the HIV proviral load have not achieved success. Knowledge
of the follicular reservoirs outlined here and the hurdles that they pose to achieving a cure could
provide some insights into these failures, as well as redirect efforts to more successful approaches.

Early attempts to activate and purge the viral reservoir were conducted using IL-2 [62] or CD3
activation [63], with the assumption that the viral reservoir would be purged from latently infected
cells through HIV cytopathic effects or immune clearance. Patients receiving IL-2 in addition to
ART had lower levels of replication-competent HIV in peripheral blood CD4+ T cells [62] but upon
interruption of ART plasma HIV levels quickly rebounded [64]. Thus, broad immune activation
and treatment cessation proved unsuccessful in purging the HIV reservoir. More recent
approaches to activate and purge the HIV reservoir have been directed at activating latent
gene expression through the use of inhibitors of histone deacetylases (HDAC). This approach
activates viral transcription without activating the cells. Clinical trials of several HDAC inhibitors
have demonstrated increases in HIV RNA expression [6567]; however, none of these studies
have revealed signicant decreases in the latent resting CD4+ T cell reservoir [6568]. One
reason for this failure has been hypothesized to be a lack of an effector CTL response, as in vitro
virus-specic CTLs have been shown to reduce latently infected cells upon activation [69]. An
alternative explanation is that virus-specic CTLs are present, but do not enter follicles, as has
been reported in untreated disease [4,6]. Thus, strategies to boost numbers of CTLs in B cell
follicles, such as by transduction of the follicular homing molecule CXCR5 into HIV-specic CTLs
[70], could be an important component of HIV cure in combination with latency-reversing agents.
Although it is unknown if this approach would have deleterious effects, such as disruption of the
GC reaction or alteration of antibody production, directing HIV-specic CTLs into the follicles
could eliminate HIV-infected cells within follicles.

The use of broadly neutralizing antibodies prophylactically after HIV exposure led to decreased
establishment of HIV reservoirs in humanized mice, and was effective in decreasing established
reservoirs when combined with a combination of viral inducers [71]. Although the humanized
mouse models are useful for studying many aspects of HIV immunopathogenesis, most do not
recapitulate key aspects of human lymphoid tissues. Specically, they lack FDCs and GCs, and
fail to develop IgG responses [72]. As noted above, FDC-bound virions are potently infectious to
TFH, even in the presence of neutralizing antibodies [13]. Thus, it would be important to carefully
examine this strategy in a more physiologically relevant model, such as the SIV-infected rhesus
macaques, before proceeding to test it in humans.

Despite the successful bone marrow transplant of the Berlin patient [61], subsequent trans-
plants have proven unsuccessful in preventing HIV rebound [73,74], although HIV DNA was not
detected in these individuals peripheral blood mononuclear cells (PBMC) prior to interruption of
therapy. Viral reservoirs also persist in simian-human immunodeciency virus (SHIV)-infected
rhesus macaques following transplantation [75]. Although some have concluded that this failure
is due to residual infection of host CD4+ T cells, it is difcult to imagine that any purging strategy
will more effectively eliminate infected CD4+ T cells than a bone marrow transplant, particularly in
cases of graft-versus-host disease, which occurred in some of these transplants. An alternative
explanation is that the transplantation regimens failed to eliminate cell-free infectious virus on
FDCs. It would be important to evaluate this possibility, particularly in animal models, and
develop approaches that are more effective against FDCs in the transplant setting. Furthermore,
strategies to displace virions bound to FDCs outside of the transplant setting should be
evaluated to determine if they diminish the reservoir. Previous work has shown the effectiveness
of complement and CD21 antibody-mediated displacement of virions from the surface of B cells

342 Trends in Microbiology, May 2016, Vol. 24, No. 5


from HIV-infected patients ex vivo [24]. An immunotoxin-conjugated agent might be effective Outstanding Questions
against FDC-bound virions as well [76]. What fraction of latently infected CD4+
T cells are memory TFH?

Concluding Remarks How long do infectious virions persist


TFH expand in number and are a major site of virus infection and replication in untreated, on FDCs in the setting of ART?
asymptomatic HIV infection as well as in treated disease. Multiple factors likely contribute to
Can removal of infectious complexes
the concentration of virus replication in TFH, including heightened permissiveness, the presence of
from FDCs reduce the HIV reservoir
highly infectious virions on FDCs, low frequencies of virus-specic CTLs in B cell follicles, and prevent viral rebound?
expansions in numbers and impairments in function. Emerging data suggest that memory TFH
constitute a signicant fraction of the latent reservoir in treated disease, and this is an important area Will expression of CXCR5 by virus-spe-
cic CTLs induce them to home to
for future research (see Outstanding Questions). Whether interactions between FDCs harboring
follicles and suppress virus replication
infectious virions and TFH further contribute to the latent HIV reservoir remains to be determined. and ultimately clear the latent HIV
Strategies aimed at targeting follicular reservoirs of HIV could provide important insights into these reservoir?
questions, and potentially a cure for HIV infection. It should be noted that, although TFH may be a
What is the relationship between follic-
major cellular reservoir, other reservoirs have been described, including the nasopharynx, lung,
ular HIV reservoirs and other HIV res-
male and female genital tracts, as well as the brain [77,78]. The relationship between follicular ervoirs throughout the body?
reservoirs and these sites, specically whether additional therapies may be necessary to achieve a
cure, remains to be determined and is an important question for future research.

References
1. Crotty, S. (2014) T follicular helper cell differentiation, function, and 16. Keele, B.F. et al. (2008) Characterization of the follicular dendritic
roles in disease. Immunity 41, 529542 cell reservoir of human immunodeciency virus type 1. J. Virol. 82,
2. Folkvord, J.M. et al. (2005) Lymphoid follicles are sites of height- 55485561
ened human immunodeciency virus type 1 (HIV-1) replication and 17. Tjernlund, A. et al. (2010) In situ detection of Gag-specic CD8+
reduced antiretroviral effector mechanisms. AIDS Res. Hum. Ret- cells in the GI tract of SIV infected Rhesus macaques. Retrovirol-
roviruses 21, 363370 ogy 7, 12
3. Perreau, M. et al. (2013) Follicular helper T cells serve as the major 18. Fukazawa, Y. et al. (2015) B cell follicle sanctuary permits persis-
CD4 T cell compartment for HIV-1 infection, replication, and tent productive simian immunodeciency virus infection in elite
production. J. Exper. Med. 210, 143156 controllers. Nat. Med. 21, 132139
4. Connick, E. et al. (2007) CTL fail to accumulate at sites of HIV-1 19. Lindqvist, M. et al. (2012) Expansion of HIV-specic T follicular
replication in lymphoid tissue. J. Immunol. 178, 69756983 helper cells in chronic HIV infection. J. Clin. Invest. 122, 3271
5. Hufert, F.T. et al. (1997) Germinal centre CD4+ T cells are an 20. Petrovas, C. et al. (2012) CD4 T follicular helper cell dynamics
important site of HIV replication in vivo. AIDS 11, 849857 during SIV infection. J. Clin. Invest. 122, 3281
6. Connick, E. et al. (2014) Compartmentalization of simian immu- 21. Hong, J.J. et al. (2012) Spatial alterations between CD4+ T
nodeciency virus replication within secondary lymphoid tissues of follicular helper, B, and CD8+ T cells during simian immunode-
rhesus macaques is linked to disease stage and inversely related ciency virus infection: T/B cell homeostasis, activation, and
to localization of virus-specic CTL. J. Immunol. 193, 56135625 potential mechanism for viral escape. J. Immunol. 188, 3247
7. Brenchley, J.M. et al. (2012) Differential infection patterns of CD4+ 3256
T cells and lymphoid tissue viral burden distinguish progressive 22. Choi, Y.S. et al. (2013) Bcl6 expressing follicular helper CD4 T cells
and nonprogressive lentiviral infections. Blood 120, 41724181 are fate committed early and have the capacity to form memory. J.
8. Thacker, T.C. et al. (2009) Follicular dendritic cells and human Immunol. 190, 40144026
immunodeciency virus type 1 transcription in CD4+ T cells. J. 23. Baumjohann, D. et al. (2013) Persistent antigen and germinal
Virol. 83, 150158 center B cells sustain T follicular helper cell responses and phe-
9. Kohler, S.L. et al. (2016) Germinal center t follicular helper cells (GC notype. Immunity 38, 596605
Tfh) are highly permissive to HIV-1 and alter their phenotype during 24. Moir, S. et al. (2000) B cells of HIV-1infected patients bind virions
virus replication. J. Immunol. Published online February 12, 2016. through CD21complement interactions and transmit infectious
http://dx.doi.org/10.4049/jimmunol.1502174 virus to activated T cells. J. Exper. Med. 192, 637646
10. Allam, A. et al. (2015) TFH cells accumulate in mucosal tissues of 25. Boswell, K.L. et al. (2014) Loss of circulating CD4 T cells with B cell
humanized-DRAG mice and are highly permissive to HIV-1. Sci. helper function during chronic HIV infection. PLoS Pathog. 10,
Rep. 5, e10443 e1003853
11. Spiegel, H. et al. (1992) Follicular dendritic cells are a major reservoir 26. Xu, H. et al. (2015) Persistent Simian immunodeciency virus
for human immunodeciency virus type 1 in lymphoid tissues facili- infection causes ultimate depletion of follicular Th cells in AIDS.
tating infection of CD4+ T-helper cells. Am. J. Pathol. 140, 15 J. Immunol. 195, 43514357
12. Haase, A.T. et al. (1996) Quantitative image analysis of HIV-1 27. Pantaleo, G. et al. (1993) HIV infection is active and progressive in
infection in lymphoid tissue. Science 274, 985989 lymphoid tissue during the clinically latent stage of disease. Nature
13. Heath, S.L. et al. (1995) Follicular dendritic cells and human 362, 355358
immunodeciency virus infectivity. Nature 377, 740744 28. Fan, J. et al. (1993) Elevated IFN-gamma and decreased IL-2 gene
14. van Nierop, K. and de Groot, C. (2002) Human follicular dendritic expression are associated with HIV infection. J. Immunol. 151,
cells: function, origin and development. Semin. Immunol. 14, 50315040
251257 29. Nakajima, K. et al. (1989) Induction of IL-6 (B cell stimulatory
15. Schacker, T. et al. (2000) Rapid accumulation of human immu- factor-2/IFN-beta 2) production by HIV. J. Immunol. 142, 531536
nodeciency virus (HIV) in lymphatic tissue reservoirs during acute 30. Harari, A. et al. (2002) Analysis of HIV-1and CMV-specic mem-
and early HIV infection: implications for timing of antiretroviral ory CD4 T-cell responses during primary and chronic infection.
therapy. J. Infect Dis. 181, 354357 Blood 100, 13811387

Trends in Microbiology, May 2016, Vol. 24, No. 5 343


31. Chowdhury, A. et al. (2015) Decreased T follicular regulatory cell/T 55. Chahroudi, A. and Silvestri, G. (2016) HIV and Tfh cells: circulating
follicular helper cell (TFH) in simian immunodeciency virus- new ideas to identify and protect. Immunity 44, 1618
infected rhesus macaques may contribute to accumulation of 56. Rodriguez-Barradas, M.C. et al. (2014) Quantitative and qualitative
TFH in chronic infection. J. Immunol. 195, 32373247 antibody responses to immunization with the pneumococcal poly-
32. Blackburn, M.J. et al. (2015) Regulatory and helper follicular T cells saccharide vaccine in HIV-Infected patients after initiation of anti-
and antibody avidity to simian immunodeciency virus glycopro- retroviral treatment: results from a randomized clinical trial. J.
tein 120. J. Immunol. 195, 32273236 Infect. Dis. 211, 17031711
33. Miles, B. et al. (2015) Follicular regulatory T cells impair follicular T 57. Sepako, E. et al. (2014) Incomplete recovery of pneumococcal
helper cells in HIV and SIV infection. Nat. Commun. 6, 8608 CD4 T cell immunity after initiation of antiretroviral therapy in HIV-
34. Colineau, L. et al. (2015) HIV-infected spleens present altered infected malawian adults. PLoS ONE 9, e100640
follicular helper T cell (Tfh) subsets and skewed B cell maturation. 58. Pallikkuth, S. et al. (2012) Impaired peripheral blood T-follicular
PLoS ONE 10, e0140978 helper cell function in HIV-infected nonresponders to the 2009
35. Haas, M.K. et al. (2015) Distinct patterns of Bcl-2 expression occur H1N1/09 vaccine. Blood 120, 985993
in R5-and X4-tropic HIV-1-producing lymphoid tissue cells 59. Cavert, W. et al. (1997) Kinetics of response in lymphoid tissues to
infected ex vivo. AIDS Res. Hum. Retroviruses 31, 298304 antiretroviral therapy of HIV-1 infection. Science 276, 960964
36. Cubas, R.A. et al. (2013) Inadequate T follicular cell help impairs B 60. Smith, B.A. et al. (2001) Persistence of infectious HIV on follicular
cell immunity during HIV infection. Nat. Med. 19, 494499 dendritic cells. J. Immunol. 166, 690696
37. Schultz and Bruce, T. et al. (2016) Circulating HIV-specic inter- 61. Htter, G. et al. (2009) Long-term control of HIV by CCR5 Delta32/
leukin-21+CD4+ T cells represent peripheral Tfh Cells with antigen- Delta32 stem-cell transplantation. N. Engl. J. Med. 360, 692698
dependent helper functions. Immunity 44, 167178 62. Chun, T-W. et al. (1999) Effect of interleukin-2 on the pool of latently
38. Chun, T-W. et al. (1997) Presence of an inducible HIV-1 latent infected, resting CD4+ T cells in HIV-1-infected patients receiving
reservoir during highly active antiretroviral therapy. Proc. Natl. highly active anti-retroviral therapy. Nat. Med. 5, 651655
Acad. Sci. U.S.A. 94, 1319313197 63. Prins, J.M. et al. (1999) Immuno-activation with anti-CD3 and
39. Finzi, D. et al. (1997) Identication of a reservoir for HIV-1 in recombinant human IL-2 in HIV-1-infected patients on potent
patients on highly active antiretroviral therapy. Science 278, antiretroviral therapy. AIDS 13, 24052410
12951300 64. Davey, R.T. et al. (1999) HIV-1 and T cell dynamics after interrup-
40. Wong, J.K. et al. (1997) Recovery of replication-competent HIV tion of highly active antiretroviral therapy (HAART) in patients with a
despite prolonged suppression of plasma viremia. Science 278, history of sustained viral suppression. Proc. Natl. Acad. Sci. U.S.A.
12911295 96, 1510915114
41. DeMaster, L.K. et al. (2015) A subset of CD4/CD8 double negative T 65. Archin, N. et al. (2012) Administration of vorinostat disrupts HIV-1
cells expresses HIV proteins in patients on ART. J. Virol. Published latency in patients on antiretroviral therapy. Nature 487, 482485
online November 4, 2015. http://dx.doi.org/10.1128/JVI.01913-15 66. Rasmussen, T.A. et al. (2014) Panobinostat, a histone deacetylase
42. Zhang, L. et al. (1999) Quantifying residual HIV-1 replication in inhibitor, for latent-virus reactivation in HIV-infected patients on
patients receiving combination antiretroviral therapy. N. Engl. J. suppressive antiretroviral therapy: a phase 1/2, single group,
Med. 340, 16051613 clinical trial. Lancet HIV 1, e13e21
43. Horiike, M. et al. (2012) Lymph nodes harbor viral reservoirs that 67. Elliott, J.H. et al. (2014) Activation of HIV transcription with short-
cause rebound of plasma viremia in SIV-infected macaques upon course vorinostat in HIV-infected patients on suppressive antire-
cessation of combined antiretroviral therapy. Virology 423, 107118 troviral therapy. PLoS Pathog. 10, e1004473
44. Rothenberger, M.K. et al. (2015) Large number of rebounding/ 68. Routy, J. et al. (2012) Valproic acid in association with highly active
founder HIV variants emerge from multifocal infection in lymphatic antiretroviral therapy for reducing systemic HIV-1 reservoirs:
tissues after treatment interruption. Proc. Natl. Acad. Sci. U.S.A. results from a multicentre randomized clinical study. HIV Med.
112, E1126E1134 13, 291296
45. Fletcher, C.V. et al. (2014) Persistent HIV-1 replication is associ- 69. Shan, L. et al. (2012) Stimulation of HIV-1-specic cytolytic T
ated with lower antiretroviral drug concentrations in lymphatic lymphocytes facilitates elimination of latent viral reservoir after virus
tissues. Proc. Natl. Acad. Sci. U.S.A. 111, 23072312 reactivation. Immunity 36, 491501
46. Gnthard, H.F. et al. (1998) Human immunodeciency virus repli- 70. Skinner, P. and Connick, E. (2014) Overcoming the immune
cation and genotypic resistance in blood and lymph nodes after a privilege of B cell follicles to cure HIV-1 infection. J. Hum. Virol.
year of potent antiretroviral therapy. J. Virol. 72, 24222428 Retrovirol. 1, 0000100003
47. Nettles, R.E. et al. (2005) Intermittent HIV-1 viremia (blips) and drug 71. Halper-Stromberg, A. et al. (2014) Broadly neutralizing antibodies
resistance in patients receiving HAART. JAMA 293, 817829 and viral inducers decrease rebound from HIV-1 latent reservoirs in
48. von Stockenstrom, S. et al. (2015) Longitudinal genetic charac- humanized mice. Cell 158, 989999
terization reveals that cell proliferation maintains persistent HIV-1 72. Karpel, M.E. et al. (2015) BLT humanized mice as a small animal
during effective HIV therapy. J. Infect. Dis. 212, 596607 model of HIV infection. Curr. Opin. Virol. 13, 7580
49. Hermankova, M. et al. (2001) HIV-1 drug resistance proles in 73. Henrich, T.J. et al. (2014) Antiretroviral-free HIV-1 remission and
children and adults with viral load of <50 copies/ml receiving viral rebound after allogeneic stem cell transplantation: report of 2
combination therapy. JAMA 286, 196207 cases. Ann. Intern. Med. 161, 319327
50. Chomont, N. et al. (2009) HIV reservoir size and persistence are 74. Cillo, A.R. et al. (2013) Plasma viremia and cellular HIV-1 DNA persist
driven by T cell survival and homeostatic proliferation. Nat. Med. despite autologous hematopoietic stem cell transplantation for HIV-
15, 893900 related lymphoma. J. Acquir. Immune Dec. Syndr. 63, 438441
51. Hale, J.S. and Ahmed, R. (2015) Memory T follicular helper CD4 T 75. Mavigner, M. et al. (2014) Persistence of virus reservoirs in ART-
cells. Front. Immunol. 6, 16 treated SHIV-infected rhesus macaques after autologous hemato-
52. Baron, B.W. et al. (1997) BCL6 can repress transcription from the poietic stem cell transplant. PLoS Pathog. 10, e1004406
human immunodeciency virus type I promoter/enhancer region. 76. Denton, A.E. et al. (2014) Fibroblastic reticular cells of the lymph
Genes Chromosomes Cancer 19, 1421 node are required for retention of resting but not activated CD8+ T
53. Kaczmarek, K. et al. (2013) T cell transcription factors and their cells. Proc. Natl. Acad. Sci. U.S.A. 111, 1213912144
impact on HIV expression. Virology Res. Treatm. 2013, 41 77. Santangelo, P.J. et al. (2015) Whole-body immunoPET reveals
54. Pallikkuth, S. et al. (2015) Peripheral T follicular helper cells are the active SIV dynamics in viremic and antiretroviral therapy-treated
major HIV reservoir within central memory CD4 T cells in peripheral macaques. Nat. Meth. 12, 427432
blood from chronic HIV infected individuals on cART. J. Virol. Pub- 78. Hellmuth, J. et al. (2015) CNS reservoirs for HIV: implications for
lished online December 16, 2015. http://dx.doi.org/10.1128/JVI. eradication. J. Virus Erad. 1, 67
02883-15

344 Trends in Microbiology, May 2016, Vol. 24, No. 5

You might also like