You are on page 1of 14

REVIEWS

Nuclear landscape of HIV1 infection


and integration
Marina Lusic1 and Robert F.Siliciano2
Abstract | To complete its life cycle, HIV1 enters the nucleus of the host cell as reverse-transcribed
viral DNA. The nucleus is a complex environment, in which chromatin is organized to support
different structural and functional aspects of cell physiology. As such, it represents a challenge for
an incoming viral genome, which needs to be integrated into cellular DNA to ensure productive
infection. Integration of the viral genome into host DNA depends on the enzymatic activity of
HIV1 integrase and involves different cellular factors that influence the selection of integration
sites. The selection of integration site has functional consequences for viral transcription, which
usually follows the integration event. However, in resting CD4+ Tcells, the viral genome can be
silenced for long periods of time, which leads to the generation of a latent reservoir of quiescent
integrated HIV1 DNA. Integration represents the only nuclear event in the viral life cycle that can
be pharmacologically targeted with current therapies, and the aspects that connect HIV1
nuclear entry to HIV1 integration and viral transcription are only beginning to be elucidated.

HIV1 infection can be controlled, but not cured, by transcriptional activity (see BOX1 for the organization
antiretroviral therapy (ART). ART suppresses HIV1 of the human genome and chromatin). Several factors
replication to undetectable levels, but, if therapy influence the selection of the target site, including the
is interrupted, viral loads rapidly rebound to pre- route of nuclear entry and cell cycle phase, chromatin
treatment levels. This is due to the formation of a reser structure and underlying sequence specificities and
voir of latently infected resting CD4+ Tcells that harbour interaction of the viral integrase with chromatin tether
integrated HIV1 and persist in all infected individuals, ing factors such as lens-epithelium-derived growth fac
regardless of the duration of ART15. Most of the provi tor (LEDGF; also known as PSIP1). The latest findings
ruses in resting CD4+ Tcells are defective and cannot confirm that the nuclear architecture also has a central
replicate, but a minority remains transcriptionally role in directing the PIC towards a permissive nuclear
competent and can cause viral load to rebound when environment that can support transcription and repli
treatment is stopped6,7. cation of the viral genome. In contrast to the actively
The life cycle of HIV1 begins with binding of the transcribed viral genome that is seen in productively
Department of Infectious viral envelope glycoproteins to receptors on the target infected cells, the integrated viral genome can undergo
1

Diseases, Integrative Virology,


cell, followed by fusion of the viral and cellular mem transcriptional silencing in resting CD4+ Tcells, leading
University Hospital Heidelberg
and German Center for brane and release of the viral core into the cytoplasm8. to the generation of a latent viral reservoir, which is a
Infection Research (DZIF), The viral RNA genome is then reverse transcribed major obstacle for the eradication of HIV1.
ImNeuenheimer Feld 324, into DNA by the viral reverse transcriptase; the newly In this Review, we discuss the integration of the HIV1
69120 Heidelberg, Germany. generated viral DNA forms a pre-integration complex genome into host cell DNA, including the influence of
2
Howard Hughes Medical
Institute; Department of
(PIC), which comprises viral DNA, the viral integrase the nuclear architecture on this step in the viral life cycle,
Medicine, Johns Hopkins and capsid proteins, and some cellular proteins. The PIC and its role in the outcome of infection. In addition, we
University School of Medicine, enters the nucleus and mediates the integration of viral will provide an overview of current and possible future
Room 879, Edward D.Miller DNA into the cellular genome (FIG.1). Nuclear entry is an approaches for the treatment of HIV1 that act on the
Research Building, 733North
active process that requires passage through the nuclear integration of, or on integrated, viral genomes.
Broadway, Baltimore,
Maryland 21205, USA. pore complex (NPC) in non-dividing cells, in which
Correspondence to M.L.
the chromosomes are separated from the cytoplasm Towards a preferred nuclear neighbourhood
marina.lusic@med.uni- by the nuclear envelope (see BOX1for the anatomy of the HIV1 can infect non-dividing cells and, hence, it enters
heidelberg.de nuclear periphery)9. Although viral integration can take the nucleus through the NPC channel. As the PIC by far
doi:10.1038/nrmicro.2016.162 place at various locations in the host cell genome, it pref exceeds the size of molecules that can pass through the
Published online 12 Dec 2016 erentially targets regions that have high gene density and NPC channel, it is generally accepted that the nuclear

NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 1



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Viral envelope glycoproteins


Surface proteoglycan proteins Maturation
that are products of the env
gene. The env gene encodes
glycoprotein 160 (gp160),
which forms a homotrimer and
is cleaved into gp120 and gp41 Assembly
by host cell proteases. The
outer glycoprotein gp120 and Receptor binding Reverse
the transmembrane protein and fusion transcription Synthesis
gp41 are embedded in the viral of viral
envelope, which forms the
proteins
Genomic
outermost layer of the virion.
RNAs
Splicing and
Viral core nuclear export
A viral component that of RNAs
contains the viral capsule Integration Transcription
protein p24, which surrounds
two single strands of HIV1
RNA, bound to the
nucleocapsid protein p7
andlate assembly protein p6. Latency
Italso contains enzymes that
are needed for the replication
of HIV1, such as reverse
transcriptase, protease, Figure 1 | An overview of the life cycle of HIV1. After fusion of the HIV1 virion with receptors on the plasma
ribonuclease and integrase,
membrane, the viral genomic RNA is reverse transcribed into DNA, followed by uncoating (disassembly of the viral capsid),
and numerous cellular proteins. Nature Reviews | Microbiology
nuclear entry and integration of the viral DNA into cellular chromatin. During productive infection, viral transcription is
Central polypurine tract
followed by splicing and nuclear export of viral RNA, and the production and assembly of new virus particles, which bud
A specific structural element from the plasma membrane and become infectious after maturation. Alternatively, integrated viral DNA can be silenced
that is involved in viral DNA through several mechanisms, thus enabling the creation of silent viral reservoirs (proviral latency).
structure and nuclear entry.
Itis formed during the
second-strand DNA synthesis import of the HIV1 PIC is an energy-dependent The matrix protein was the first component of the
of reverse transcription as a active process that is governed by different viral and PIC proposed to have a role in nuclear import in non-
central 99nucleotide-long cellularfactors. dividing cells as it has a nuclear-localization signal (NLS)17.
plus-strand overlap in the
linear DNA molecule; it is
However, the role of matrix in nuclear import seems to
located centrally in the Viral factors. Several viral components, including the be redundant, because even without a functional matrix
genome, in the integrase capsid, matrix, Vpr, integrase and the central polypurine HIV1 can still infect macrophages18.
openreading frame, and acts tract, have been reported to have a role in nuclear import Vpr, which is a small viral protein that is best known
as a cis determinant of
of the HIV1 PIC9,10 (FIG.2). for its capacity to arrest the cell cycle in the G2M phase,
lentiviral DNA nuclear import.
The capsid protein is a major determinant for also contains an NLS, although a non-canonical one, and
Twolong terminal repeat nuclear entry and for the infection of non-dividing it enters the nucleus in a non-classical way through its
circles cells. This was first determined when the replication interaction with importin19. Vpr was among the first
(2LTR circles). Unintegrated, of a chimeric HIV1 virus that carried the capsid pro viral proteins that were shown to be involved in the dock
circular molecules of viral
DNAthat contain two adjacent
tein of murine leukaemia virus was shown to depend ing of the PIC to the NPC and binding to Nups20,21. As
LTR promoter regions. They on the cell cycle. Arrest of the cell cycle caused a defect Vpr can move between the cytoplasm and the nucleus, it
are considered byproducts in nuclear import of this chimeric virus, as measured was suggested that Vpr has a role in the transport of viral
of integration, and their by the decreased formation of two-long terminal repeat DNA19. Furthermore, GFP-tagged Vpr was used to track
quantification by PCR-based
circles (2LTR circles)11. Furthermore, certain HIV1 the trafficking of PICs along cytoplasmic microtubule
methods is used as an
indicator of the efficiency of capsid mutants are defective only in resting cells and filaments towards the nucleus22. However, the role of Vpr
nuclear import. not in actively dividing cells, which highlights the role in nuclear import remainscontroversial.
of the capsid in nuclear entry 12. The capsid protein
Nuclear-localization signal interacts with RANBP2 (also known as NUP358) to Cellular factors. Various host proteins have been shown
(NLS). A motif that is present
inproteins that are imported
dock the virion to the cytoplasmic side of the NPC13,14, to participate in HIV1 nuclear import, many of which
into the nucleus by importins which supports the hypothesis that viral uncoating were revealed in genome-wide short interfering RNA
and adaptor proteins, which and the completion of reverse transcription occur at (siRNA) screens; those identified in more than one
interact with the nuclear the nuclear pore (reviewed in REF.10). A recent study screen include RANBP2, transportin 3 (TNPO3: also
porecomplex.
in primary macrophages showed that, intriguingly, known as TRN-SR) and NUP153 (REFS2325). Knock
Phe-Gly repeats the capsid remains associated with nuclear PICs 15, down of these factors blocks infection after reverse tran
(FG repeats). which indicates that it has a role in the nuclear scription, but before integration, and involves the capsid,
Phenylalanine-rich and portion of the viral life cycle as well. In addition to although the exact mechanisms remain to be elucidated.
glycine-rich domains found in nucleoporins (Nups), the capsid of HIV1 interacts RANBP2 and NUP153 localize to the cytoplasmic
nucleoporins that facilitate the
transport of cargo through the
with other cellular factors; disrupting these inter and the nuclear ends of the NPC, respectively, and
channel of the nuclear pore actions through mutation can substantially alter the both are attachment sites for molecules that traverse
complex (NPC). integration profile14,16. the NPC. Both proteins are enriched in Phe-Gly repeats

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | The mammalian nucleus


Nuclear architecture and genome organization
Different levels of organization of chromatin structure the nucleus, with chromosome territories being the major
organizing units. The packaging of DNA into the nucleus results in physical interactions between loci, even between
those located on remote parts of chromosomes. These interactions are non-random. High-resolution chromatin mapping,
which is based on chromosome conformation capture (3C), and related methods, showed that recurrent and functionally
significant interactions organize the genome.
Long-range interactions on the same chromosome (intrachromosomal) can occur between promoter and terminator
regions of a gene, as well as between enhancers and promoter regions. These interactions, together with those that are
reported to be driven by insulators, contribute to the organization of each chromosome into distinct structural (and
possibly functional) domains of different length. Topologically associated domains (TADs), which are separated by
genetically defined boundary elements, vary in length from several hundred bases to several million bases. Two regions
in one TAD interact with each other more frequently than with regions outside of the TAD, which means that TADs
represent the physical compartmentalization of the genome. TADs have two basic features: self-association of regions
in TADs and insulation towards neighbouring TADs. The nature of TADs is hierarchical, with sub-TADs, loops and
insulating neighbourhoods representing the local organization of each TAD. Whereas TADs seem to be conserved
between different cell types, sub-TADs, loops and insulating neighbourhoods can differ. Thus, TADs could represent a
larger, invariant feature of genome organization, whereas the underlying hierarchical structures may have roles in
lineage-specific genome regulation. Other chromatin domains that are defined on the basis of their interaction with
thenuclear lamina (lamin-associated domains (LADs)) correlate with low gene expression and late replication timing.
LADs that are present in all cell types analysed thus far are termed constitutive LADs, whereas those that differ between
cell types or change in their association with the nuclear lamina, which is dependent on the cell cycle, are known as
facultative LADs. Changes in the association with the nuclear lamina are a further indication of how dynamic
nuclearorganization really is, not only in terms of differentiation, but also in response to different exogenous stimuli,
such as viral infection (see REFS164166 for comprehensive and detailed reviews on nuclear structure and organization).
The nuclear periphery and implications on genome organization and function
Chromatin domains that have different transcriptional and replicative activities define the nuclear architecture.
Organization occurs at various levels, ranging from chromosome territories and sub-chromosomal organizational
domains to functional nuclear compartments, such as transcription or replication factories, or nuclear bodies
(Polycombbodies, promyelocytic leukaemia (PML) bodies or Cajal bodies)164,165,167.
The nuclear periphery contains the nuclear envelope, the underlying nuclear lamina and nuclear pore complexes (NPCs),
which span from the cytosol into the nucleoplasm. The nuclear envelope consists of two membrane systems, theouter
nuclear membrane (ONM), which faces the cytosol, and the inner nuclear membrane (INM), which surrounds the
nucleoplasm. These two layers are separated by the perinuclear space and intersect at the level of NPCs. The inner
side of the nuclear envelope is covered with lamins, which form a thin protein layer (2030nm) that is tightly associated
with the INM on one side and with the underlying chromatin on the other side. Type A and type B lamins, together with
lamin-associated proteins, constitute lamin filaments and provide structural support to the nucleus. The key function
of nuclear lamin proteins is to maintain the shape and mechanical properties of the nucleus; they also tether
heterochromatic chromosomal domains to the nuclear periphery. In the nuclear lamina, genes are mostly transcriptionally
inactive and this region is enriched in heterochromatin111,168,169, whereas there is no condensed chromatin beneath the NPC170.
The mammalian NPC is composed of 30 different nucleoporins (Nups). Nups are present in multiple copies and
arranged in a ~125nm diameter core that can be structurally divided into a core scaffold (central pore), which is formed
by NUP93 and NUP205, and two rings (the cytoplasmic ring and the nuclear ring) that are composed of NUP107 and
NUP160. The inner side of this structure contains the socalled phenylalanine-glycine Nups (FGNups), such as NUP62,
which regulate receptor-mediated transport through the nuclear pore. Embedded in the nuclear envelope are three
known transmembrane Nups (POM121, NDC1 and GP210 (also known as NUP210)), that link the NPC to the nuclear
envelope. On the cytoplasmic side, eight filaments (formed of NUP214, RANBP2 and NUP88) protrude towards the
protein synthesis machinery and the cytoskeleton. Nuclear basket filaments that are composed of NUP153, TPR and
NUP50 (REFS112,113) have a more direct role in genome-related functions, whereas all other components of the nuclear
pore participate in selective nuclear import and export.

(FG repeats) and establish contacts with nuclear recep could dock HIV1 to the cytoplasmic side of the NPC.
tors. They have homologous zinc-finger domains that Nuclear import most likely occurs in synergy with
could confer their binding to DNA (or RNA) and could other Nups, such as NUP153 (REF.14), as the depletion
also explain their capacity to influence target site pref of RANBP2 or NUP153 decreases the nuclear import of
erences for integration14,16. In fact, NUP153 has been HIV1 (REFS14,24,26,29). CYPA binding and cis-to-trans
shown to affect integration site distribution16 and nuclear isomerization lead to conformational changes in the
import of HIV1 through its interaction with the capsid capsid; however, the effect of these processes on
Zinc-finger domains protein26,27. the uncoating and infectivity of HIV1 remain poorly
Protein structural domains that Cyclophilin A (CYPA) is a peptidyl-prolyl isomer understood30.
can coordinate one or more
zinc atoms to act as binding
ase that binds to an exposed loop on the surface of TNPO3 was also identified in siRNA screens23 and
partners for a wide variety of capsid28. It was suggested that CYPA directs HIV1 into an independent yeast two-hybrid screen showed its
substrates, including DNA. a nuclear entry pathway that involves RANBP2, which interaction with integrase31. TNPO3 imports splicing

NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 3



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

capsid protein (N74D) has been shown to be resistant to


CPSF61358. This mutant is insensitive to TNPO3 knock
down, whereas the wild-type CPSF6 changes its locali
zation from the nucleus to the cytosol when TNPO3 is
depleted38. It was proposed that CPSF6 enables HIV1 to
use RANBP2, NUP153 and TNPO3 for nuclear entry 39
and the mutation in the CPSF6 binding site of capsid
(N74D) was shown to activate alternative mechanisms for
the PIC to enter the nucleus16,29. This ultimately results in
Reverse integration into gene-poor regions14,16. A recent study has
transcription provided compelling evidence that CPSF6 gene silencing
Capsid results in HIV1 integration in sites distinct from gene
CYPA
bodies and gene-dense chromatin regions40 (see below).
Vpr
Nuclear docking and uncoating of the virion are
Matrix Integrase tightly connected with the import of the PIC into the
vDNA nucleus and are dependent on the capsid, which enters
the nucleus together with the viral DNA14,15,36,41. Thus
nuclear pore proteins from both the cytosolic and nuclear
RANBB2 sides of the NPC, as well as other transport proteins,
determine viral entry from the cytosol into the nucleus
and towards the susceptible regions of the genome in
TNPO3 which it will integrate.
Nuclear basket
CPSF6 (including NUP153
and TPR)
HIV1 integration
After passing through the NPC, the PIC reaches the
periphery of the nucleus, which is a complex and hetero
geneous environment that provides anchoring sites for
chromosomes.
Capsid
Integrase LEDGF Integrase. Insertion of the viral DNA into chromosomes
is catalysed by integrase42,43 (FIG.3a). Integrase is encoded
5 3 by the pol gene, and the mature enzyme, which is com
posed of three structurally and functionally distinct
3 5 domains4446, is derived from the GagPol polyprotein
Target DNA precursor following cleavage by the viral protease42.
The aminoterminal zinc-binding domain (NTD)
Figure 2 | Nuclear entry and the selection of integration site. The capsid of of integrase consists of three -helices that contrib
HIV1 seems to be a crucial viral factor for both uncoating Nature
and theReviews | Microbiology
entry of the ute to oligomerization and to the catalytic activity 47,48.
pre-integration complex (PIC) into the nucleus. Other than capsid, the viral factors Vpr Integrase also contains a conserved HHCC motif,
and matrix, as well as different cellular partners are involved in the process of nuclear which comprises two histidine and two cysteine resi
entry on the cytosolic side of the nuclear pore complex (NPC). Cyclophilin A (CYPA; dues. This domain binds to a Zn2+ ion and resembles the
also known as PPIA) and RANBP2 have roles in the nuclear entry of the PIC on the zinc-coordinating residues of zinc fingers47.
cytosolic side of the NPC, transportin 3 (TNPO3) is involved in shuttling through the The central catalytic core domain (CCD) is highly con
NPC, and nucleoporin 153 (NUP153) and cleavage and polyadenylation specificity served in all retroviral integrase proteins, and, in addition
factor 6 (CPSF6) are involved in the import of HIV1 into the nucleus. Both NUP153 and
to its catalytic activity, has been proposed to be involved
CPSF6 have a role in target site selection, together with lens-epithelium-derived
growth factor (LEDGF), which is the most prominent chromatin-tethering factor that is
in DNA binding 44. From a structural perspective, the
involved in HIV1 integration. vDNA, viral DNA. catalytic core domain belongs to the RNaseH superfamily
of polynucleotidyl transferases, and its key active site46 is
comprised of three acidic amino acid residues (Asp64,
factors into the nucleus32 and was suggested to pro Asp116 and Glu152) with a conserved spacing of 35 amino
mote the nuclear entry of the PIC13,14,31. The same role acids between Asp116 and Glu152 (D,D(35)E motif).
was attributed to another cellular factor, cleavage and These residues form coordination bonds with two Mg 2+
polyadenylation specificity factor 6 (CPSF6), that was ions during nucleophilic substitution and DNA strand
shown to interact with the capsid protein before trans transfer 4446. The CCD interacts with both the donor
location into the nucleus3336. CPSF6 is a 68kDa member (viral) and the target (cellular) DNA, and mutations in the
of a large protein complex of pre-mRNA splicing factors D,D(35)E motif inhibit integrase activity 49,50. The CCD
that shuttle between nucleus and cytoplasm37. Atrun also contains a serine residue that can be phosphorylated
cated form of CPSF6, lacking the carboxy-terminal by cellular JUN Nterminal kinases and subsequently
domain (CPSF61358), that localized mainly in the cyto isomerized by peptidyl-prolyl cis-trans isomerase
sol and blocked nuclear entry of the PIC, was identi NIMA-interacting 1 (PIN1); this post-translational
fied in a mouse cDNA screen13. A mutant form of the modification stabilizes integrase in Tcells51.

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Pol Figure 3 | Integrase and the integration mechanism.


a | Schematic representation of HIV1 integrase, which is
Gag PRO RT IN
cleaved from the GagPol protein: integrase domains are
shown in the lower part of the panel with numbers
representing amino-acid position boundaries and the
Primary lengths of interdomains and the carboxyterminal
destabilizing Protease tail. Amino acid residues that are conserved between
residue cleavage site
different integrase species are marked by red stars; point
mutations in these residues abolish the activity of the
1 47 59 202 223 270 enzyme. Prototype foamy virus (PFV) integrase, which was
HIV-1 Phe NTD CCD CTD used for the crystal structure of the intasome, is also shown.
D64 D116 E152 288 b | HIV1 integration mechanisms: integrase multimerizes
1 4851 102 123 271 321 374 392 on the ends of the viral long-terminal repeat (LTR) and then
PFV
cleaves two nucleotides from each 3 end of the viral DNA
during 3 processing. The completion of 3 processing
leaves a recessed and chemically reactive hydroxyl group.
b Following nuclear import of the pre-integration complex
IN (PIC), integrase binds to the target DNA and strand transfer
IN occurs. After disassembly of the strand transfer complex,
LTR LTR the DNA recombination intermediate is repaired, probably
HIV DNA
Intasome formation by host cell enzymes, and the provirus, flanked by host
IN DNA, becomes an integrated part of the cellular chromatin.
IN CCD, catalytic core domain; CTD, carboxy-terminal
domain; IN, integrase; NTD, amino-terminal domain;
PRO, viral protease; RT, reverse transcriptase.
3 processing
CAGT-3 5-ACTG
GTCA-5 3-TGAC
The partial structures of these domains, resolved
LTR LTR
either by crystallization46 or by NMR spectroscopy53,
were useful for proposing different models of integrase
in complex with the ends of viral DNA55. However, full
Dissassembly of coverage of invivo events became available only after
strand transfer complex the crystal structure of prototype foamy virus (PFV)
CA-OH-3 5-ACTG
LTR GTCA-5 3-HO-AC LTR integrase in complex with viral DNA was resolved56,57.

Molecular mechanisms of integration. Integrase carries


out two essential enzymatic reactions: 3-end processing
of the viral DNA and strand transfer reactions (FIG.3b).
Both 3-end processing and the integration of oligo
nucleotides that match the ends of HIV1 DNA can be
reproduced invitro in the presence of integrase alone58,59.
Moreover, experiments that used hybrid viruses with
altered integration preferences supported the principal
role of integrase in the selection of the integrationsite60.
Integrase activity initiates soon after the synthesis
of a double-stranded viral DNA molecule. In the PIC,
5 Gap repair and 3 integrase multimerizes on the nascent DNA to form the
integration into
3 cellular genome
5 functional integraseviral DNA complex known as
the intasome56,57. As revealed by the Xray crystal struc
ture of integrase from PFV, the intasome consists of a
tetramer (or dimerofdimers) of integrase subunits56,57,
5 3 and integrase activity depends on tetramerisation61. One
3 5 subunit of each integrase dimer binds to viral DNA for
Nature Reviews | Microbiology 3-end processing: integrase removes two nucleotides
from 3 ends of the blunt-ended linear viral DNA and
The Cterminal domain (CTD) has the least generates reactive CAOH3-hydroxyl ends.
sequence conservation among retroviral integrase pro After import into the cell nucleus, the intasome docks
teins52, with residues 220270 being responsible for onto the target DNA to form a target capture complex
DNA binding and the formation of dimers of parallel (TCC)56 and strand transfer ensues: integrase uses the
monomers53. The CTD contains four lysine residues 3hydroxyl groups on the ends of the viral DNA as
that are acetylated by cellular histone acetyltrans nucleophiles to attack the target DNA across the major
ferases. These post-translational modifications regulate groove. Complementary strands of viral DNA are then
integrase strand transfer activity, viral integration and joined with the 5phosphates at the ends of the target
ultimately result in decreased viral replication54. DNA. The resulting DNA recombination intermediate

NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 5



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

contains single-stranded gaps at the joints that can be This preference was then confirmed invivo in resting
completed by the cellular repair machinery. This yields CD4+ Tcells from infected individuals70. Subsequently,
a 5bp target site duplication that flanks the stably inte a detailed map of preferred target sites was generated,
grated provirus62. Modelling that was based on the crys which confirmed that in most cell lines HIV1 integrates
tal structure of the PFV intasome suggested that HIV1 into transcriptionally active regions60,7376. HIV1 integra
integrase accommodates target DNA with a severely bent tion occurs in active transcriptional units72,77,78. A recent
conformation; integrasetarget DNA interactions prefer study of approximately 1 million integration sites in
specific, flexible dinucleotides at the centre of integra infected HEK293 cells showed that 75% of integrations
tion sites63,64. Several amino acids of PFV integrase were occurred in active, RNA pol II-dependent transcriptional
shown to distort the target DNA57. Homology modelling units that had numerous introns; when corrected for
between the TTC of HIV1 and the crystal structure of their relative length, no selection of intronic over exonic
the PFV intasome showed that the amino acids Ser119 sequences was observed40. Analysis of 1,000 transcrip
and Arg231 of the HIV1 integrase are in direct contact tional units that had the largest number of total integra
with the target DNA65,66. Careful invitro analysis of 13 tions revealed that integration density correlates strongly
integrase mutations yielded a total of 1610 unique inte with the levels of splicing, and that the cellular protein
gration sites, from which a preferred target DNA signa LEDGF is required for targeting highly spliced transcrip
ture emerged. HIV1 targets (0)RYXRY(4) sequences tional units, through its direct interaction with numerous
(in which R refers to a purine and Y refers to a pyrim splicing factors79. Large sets of data on the integration
idine), which favours overlapping flexible dinucleo sites of primary Tcells from individuals infected with
tides at the centre of the integration site; whether this HIV1 confirmed that HIV1 preferentially integrates
sequence is the one that is responsible for the preferred into highly transcribed genes7,80,81. Interestingly, these
targeting of gene-dense regions still remains to be deter recent studies have provided strong evidence that inte
mined65. Although the preference for integration into this gration into particular genes can cause clonal expansion
sequence is weak, it still predicts integration targeting well and persistence of the infected cell7,8082. Analysis of patient-
when compared with randomly chosen sequences65,67,68. derived samples demonstrated that HIV1 shows a strong
Interestingly, polymorphisms at position 119 and posi preference towards expressed genes6,74,83 with several
tion 231, which are frequently observed, can re-target the independent integrations identified in certain genes. One
intasome away from transcriptionally active regions66. In study 81 identified 15 independent integrations in intron4
particular, two variants (S119G and R231G) retain cata and intron 5 of the BACH2 gene, all of which were in the
lytic activity while having altered requirements for target same transcriptional orientation as the host gene. By con
DNA binding and decreased electrostatic compatibility trast, invitro infection of HeLa cells or haematopoietic
with certain nucleosomes. Consequently, integration was stem cells showed integrations throughout the gene in
distorted away from gene-dense chromatin regions; these either orientation. The MKL/myocardin-like2 (MKL2)
polymorphisms were also proposed to accelerate disease gene also showed this interesting pattern of multiple
progression in longitudinal follow-updata66. invivo integrations in a specific region of the gene in the
A single-particle cryo-electron microscopy study same transcriptional orientation76. Another study iden
demonstrated how the PFV intasome captures chromo tified BACH2 integrations in two out of three patients80.
somal DNA that is wrapped around a mononucleosome. BACH2, signal transducer and activator of transcrip
During this interaction, the histone octamer remains tion5B (STAT5B) and MKL2 are recurrent integration
intact while DNA is lifted from the H2AH2B surface to sites for HIV1, with each site identified multiple times in
enable integration at a strongly preferred location. In the different patients7,80,81,83. Interestingly, these genes encode
case of HIV1, this occurs preferentially at 3.5 turns of cellular transcription factors that are involved in cell
the DNA superhelix. On the basis of mutagenesis studies growth, with BACH2 and MKL2 being reported to have
that changed the intasome preferences for chromatin, the proto-oncogenic functions84,85. Frequent identification of
authors proposed that contacts between the intasome and these integration sites are a result of the clonal expan
the Nterminal tails and Cterminal domains of histones sion of Tcells after HIV1 integration, which means that
enable the intasome to decode epigenetic marks69,70. HIV1 integration into these genes increases the long-
term survival of the host cells and possibly also alters the
Finding the right target function of thesegenes.
Integration can hypothetically occur throughout the Many genes that were identified in patient stud
genome; nevertheless, it is not a random process71. HIV1 ies7,80,81,83 are also found in invitro HIV1 infections of
integrase prefers a specific sequence at the end of the viral primary CD4+ cells, peripheral blood mononuclear
DNA, whereas at the chromatin level the selection of inte cells (PBMCs) or Tcell lines68,72,86,87. These genes, which
gration site is influenced by three major determinants: reappear in different data sets, termed recurrent integra
Nucleosomes sequence specificity, chromatin structure and cellular tion genes (RIGs), have common features: they are local
The building blocks of tethering factors. ized in specific regions of chromosomes, as previously also
chromatin, which are suggested for the so-called hotspots of integration72, and
composed of an octameric Sequence specificity and chromatin determinants. are positioned in the outer shell of the Tcell nucleus87.
histone core around which
147bp of DNA are wrapped
HIV1 preferentially integrates into active genes. The Considering that integration into some of these genes
(in1.65 turns of a left-handed first integration site mapping showed that HIV1 favoured may lead to clonal expansion of the target cells, further
superhelix). integration into transcriptional units in a Tcell line72. analysis of their genetic architecture and transcriptional

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Euchromatin capacity is required. This is of particular importance Other factors besides LEDGF are also involved in
A loosely packed form of because the clonal expansion of infected cells that carry chromatin tethering and integration site selection and
chromatin (DNA, RNA and replication-competent HIV1 could greatly complicate some of them are included in the PIC before nuclear
histones) that is usually efforts to eradicate the infection. import. High mobility group protein A1 (HMGA1) was
enriched in genes that are
often being actively
Integration into transcriptional units correlates among the first proteins to be identified as an integration-
transcribed. Most of the with several features, such as high GC content, high relevant factor 104 . This non-histone chromatin-
genome is euchromatic CpG island density and specific histone marks; indeed, associated protein can interact with viral DNA, but
(predicted at approximately euchromatin can easily be accessed by the viral PICs40,88. not with integrase, and might help in bringing the
90%), whereas the rest is
Invivo, HIV1 integration favours the major groove viral DNA ends together in the intasome104. Barrier-to
heterochromatic, that is,
tightlypacked and not
of DNA, facing outwards from the nucleosome core77. autointegration factor (BAF), a protein that is involved in
activelytranscribing. Analysis of approximately 40,000 integration sites in a the assembly and organization of the nuclear envelope,
Euchromatin is often referred Tcell line revealed associations between integration is included in PICs, in which it interacts with viral DNA
to as open chromatin. frequency and transcription-associated histone mod probably to compact it and prevent premature self-inte
SWI/SNF chromatin
ifications77. For example, H3K9 acetylation, which is gration9. Other cellular factors can interact with integrase
remodelling complex characteristic of transcriptionally active genes, and directly, such as integrase interactor 1 (INI1). Although
A complex that remodels trimethylated H3Lys36 (H3K36me3), which is associ the importance of INI1 in the stimulation of HIV1 inte
nucleosomes using the ated with elongating RNA pol II and productive tran gration is debated, INI1 could, as part of the SWI/SNF chro
hydrolysis of ATP to regulate
scription, correlated with HIV1 integration sites87. matin remodelling complex, facilitate targeted integration
the accessibility of DNA to the
transcription machinery.
Importantly, LEDGF, the main chromatin-tethering into SWI/SNF-remodelled regions of the genome105.
factor of HIV1, binds to H3K36me3. Several cellular partners that are able to post-
Other retroviruses also show high preferences for translationally modify HIV1 integrase have also been
euchromatin, but usually target promoter regions89, reported to affect its enzymatic activity. In particular,
whereas HIV1 generally integrates into transcriptional acetylation by histone acetyl transferase (HAT) p300
units and introns, which indicates that factors that are increases the affinity of integrase for DNA54. Acetylated
unrelated to chromatin also contribute to the selection integrase can be bound by transcription intermediary
of integrationsite. factor 1 (TIF1, also known as TRIM28 and KAP1), a
member of the tripartite motif (TRIM) family of antiviral
Chromatin tethering. LEDGF is probably the most proteins that, in a complex with histone deacetylase1
important host factor for HIV1 integration, determining (HDAC1), deacetylates integrase and restricts integra
both integration efficiency and integration site selection. tion106. It is intriguing to hypothesize that the binding of
LEDGF is a chromatin-tethering factor that has been TIF1 to integrase could serve to re-target the viral genome
implicated in cellular differentiation and response to towards the more repressed regions of cellular chromatin
stress90,91. It was initially identified as an integrase cofac in which the virus could remain silent for certain periods
tor 92 by coimmunoprecipitation; numerous studies that of time; however, this requires further exploration.
followed have confirmed its pivotal role in the replication
of HIV1 (REFS9397) (reviewed in REF.98). The full-length Determinants of HIV1 positioning in the nuclear space.
isoform of 75kDa contains a Cterminal integrase- Recent evidence that CPSF6 has a role in integration site
binding domain (IBD), which is absent in the trun targeting has focused attention on the role of capsid.
cated isoform of 52kDa; both isoforms contain the CPSF6, which binds to capsid rather than HIV1 inte
conserved Nterminal PWWP domain (Pro-Trp-Trp-Pro), grase, was shown to markedly influence integration into
and both were shown to associate with several splic transcriptionally active spliced genes and regions of open
ing factors79,99. The IBD tethers integrase to chromatin chromatin40. A tandem knockout of CPSF6 and LEDGF
through the conserved Nterminal PWWP domain, decreased integration into genes and revealed that CPSF6
which binds specifically to H3K36me3 (REFS99,100). had a more dominant role than LEDGF in targeting
The association of LEDGF with active regions of chro HIV1 integration to euchromatin. A two-phase mecha
matin, which was first probed by DNA adenine methyl nism, in which capsidCPSF6 binding directs HIV1 to
transferase identification (DamID)101,102, most likely actively transcribed chromatin and integraseLEDGF
occurs through a direct interaction with splicing fac binding directs integration into gene bodies, seems a
tors79,99. LEDGF thus directs the integration of HIV1 very probable explanation; this reflects the complex
into highly spliced transcriptional units and intron-rich interplay between the incoming PICs and the host cell
genes79. Depletion of LEDGF shifts integration away chromatin. This hypothesis is supported by data from
from active genes93,97. Chimeric LEDGF molecules, in a recent independent study in which the spatial nuclear
which the chromatin-binding module was replaced by distribution of HIV1 PICs was assessed in the presence
the chromatin-reader domains of other proteins, also or absence of LEDGF; it was shown that HIV1 maintains
showed a shift in integration patterns78,103. Hepatoma- its spatial distribution in the euchromatic regions of the
derived growth factor-related protein 2 (HDGFRP2), a genome independent of LEDGF107, which suggests that it
host protein that also has PWWP and IBD domains, is is indeed the capsidCPSF6 interaction that directs the
often associated with the residual integration capacity of PIC towards the preferred regions in the nucleus.
HIV1 in LEDGF knockdown cells; however, studies with The nucleus is a highly dynamic and compart
cells that lack both LEDGF and HDGFRP2 have shown mentalized environment, in which chromosomes and
residual, non-random integration in active genes96. genes occupy preferred positions108; this high degree

NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 7



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Nuclear basket of the HIV1 genome. Especially interesting is the role of


Nuclear (including NUP153, Nuclear pore complex TPR, a protein of the inner basket of the NPC. TPR
lamina NUP98 and TPR) knockdown has almost no effect on the efficiency of viral
integration (as measured by Alu-PCR), but markedly
Nuclear envelope
impairs viral transcription87,88,114, which is consistent with
the decreased deposition of open chromatin marks, in
particular H3K36me3 (which associates with the elongat
ing RNA pol II), on HIV1 target genes114. TPR regulates
LAD the expansion of heterochromatic regions in proximity
CPSF6
to the NPC115. Furthermore, TPR stabilizes LEDGF in the
HIV-1 nuclear periphery 114, which implies that the two proteins
LEDGF
might work together to direct HIV1 DNA towards its
Open preferred sites in the nuclearperiphery 116.
chromatin
Higher rate of HIV-1 Functional implications of integration
1 m integration
Some aspects of the positioning of integration sites seem
Lower rate of HIV-1
to be a consequence of nuclear entry through the nuclear
integration pore. An example is the involvement of NUP153 in the
distribution of integration sites16,26,27. Moreover, the pro
moter activity of integrated LTR promoters depends
on an open chromatin conformation and the binding
of different transcription factors117,118. Thus, it is highly
Figure 4 | HIV1 integration at the nuclear periphery. HIV1 integration
Nature Reviews into
| Microbiology probable that if regions of open chromatin are in close
transcriptionally active genes and regions of open chromatin is enabled by the cellular
proximity to nuclear pores, they will be preferred sites
factors lens-epithelium-derived growth factor (LEDGF), which is a chromatin-tethering
factor that binds to HIV1 integrase, and cleavage and polyadenylation specificity factor6 for integration and viral gene expression.
(CPSF6), which binds to the capsid protein. HIV1 integration, which occurs in the outer The functional implications of integration site posi
shell of the nucleus in proximity to the nuclear pore complex (NPC), is also dependent tioning in the human genome are not fully understood,
onnucleoporin 153 (NUP153), NUP98 and TPR. Consistently, highly targeted HIV1 despite several attempts to address this question experi
integration sites (or genes) are preferentially positioned in the outer shell of the nucleus, mentally. It still remains unclear how strong the relation
1m beneath the nuclear envelope. LAD, lamin-associated domain. ship is between the selection of integration site and the
necessity for the provirus to be efficiently and rapidly
transcribed (and continue with the productive life cycle).
of spatial organization determines nuclear functions109. The spatial positioning of the viral genome in proximity
Visualization of infected primary human CD4+ Tcells of the nuclear pore is depicted in FIG.5a. Similarly, it is still
by 3D immuno-DNA fluorescence insitu hybridiza unclear whether and how the integration sites relate to
tion revealed that HIV1 preferentially integrates in the the capacity of the provirus to establish a latent infection.
peripheral nuclear compartment, within 1m from Following integration into the cellular genome, the fate of
the edge of the nucleus87. This is consistent with previ the provirus is determined by several factors. Active tran
ous findings that showed preferential peripheral nuclear scription of the provirus leads to the production of new
localization of viral PICs. PICs, visualized either by viral progeny, whereas silencing, which is caused by sev
EGFP (enhanced green fluorescent protein) fused to eral factors, leads to transcriptional repression and pro
integrase88,107 or by YFP (yellow fluorescent protein) viral latency 119. Among different factors that contribute
fused to the host protein APOBEC3F110, were distrib to latency, HIV1 chromatin structure as well as provirus
uted in close proximity to the nuclear envelope and not location in the host chromatin depend on the integra
throughout the nucleus. tion site. Taking into account preferential integration into
The nuclear architecture could thus determine the open chromatin regions, it remains to be determined how
selection of integration site (FIG.4); in fact, highly targeted the heterochromatic status of the provirus is established
RIGs were observed to be prevalently distributed at the during latency (FIG.5b,c shows some options).
nuclear periphery 87. RIGs are characterized by the pres One possibility is that the virus is initially integrated
ence of RNA polymerase II and association with open into permissive regions of the genome, but then becomes
chromatin marks; by contrast, HIV1 strongly disfa repressed during the transition of the target cell from an
vours the heterochromatic condensed regions in lamin- activated state towards a quiescent state through direct
associated domains (LADs)111 as well as other centrally effects such as repressive chromatin marks deposited
located transcriptionally active regions87. The nuclear on the sites of integration120122, CpG promoter methyla
periphery contains open chromatin regions that under tion123,124, decreased binding of transcription factors some
lie the NPC (comprehensively reviewedin REF.112), and of which are excluded from the nucleus in resting CD4+
HIV1 DNA is bound by several proteins of the NPC87. Tcells125127, and transcriptional interference from neigh
Nups have important roles in cellular functions such as bouring host genes128130 (reviewed in REFS117,119,131).
nuclear trafficking, chromosome segregation during Silencing of the HIV1 genome through an association
mitosis and genome integrity 112,113. Tethering to the NPC with alphoid pericentromeric regions from a different
could be required for the optimal transcriptional activity chromosome132 suggests that trans-acting mechanisms

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Figure 5 | Connecting integration and transcription.


Nuclear pore Nuclear basket
complex (including NUP153 a|HIV1 integration, which is mediated by the cellular
Nuclear lamina and TPR) factors lens-epithelium-derived growth factor (LEDGF)
and cleavage and polyadenylation specificity factor 6
Nuclear envelope (CPSF6), occurs in proximity to the nuclear pore complex
(NPC), where the association of the viral genome with
nucleoporin 153 (NUP153) and TPR facilitates viral gene
expression. The surrounding chromatin environment is
defined as euchromatic (green). Heterochromatic regions
are in orange. b | Changes in cellular activation state could
trigger the modification of chromatin organization,
CPSF6 resulting in a heterochromatic environment surrounding
the integrated HIV1 genome. This can occur following
LEDGF integration of the viral genome in the nuclear periphery.
Association with the nuclear lamina, in which histone
and DNA-methylation patterns contribute to the
heterochromatic environment, could make the viral genome
less accessible to transcription factors. c|Alternatively,
HIV1 can integrate in the inner regions of the nucleus where
it becomes silent upon encountering different nuclear
subcompartments (such as promyelocytic leukaemia (PML)
nuclear bodies or other heterochromatic regions).
b

could also be involved in latency. In fact, the association of


transcriptionally inert HIV1 with promyelocytic leukaemia
bodies (PML bodies) that was recently described as an
example of silencing in trans118,133, adds to the importance
of the spatial organization of the nucleus in determining
the fate of HIV1 (FIG.5c).
Silencing of the viral genome could also occur
through direct integration into heterochromatic
regions, which would not be permissive for transcrip
tion. Genomic location of integration was first pro
posed to contribute to latency when it was shown in a
cell line model that HIV1 transcriptional control is, in
part, dependent on the state of the surrounding chro
matin134. Regulation of HIV1 transcription was shown
to be dependent on the orientation of the integrated
viral genome and on host gene transcriptional read-
c through128,129. However, a comprehensive meta-analysis76
of integration sites in five well-characterized models of
HIV1 latency in combination with known transcrip
tional regulatory genomic features (such as histone
acetylation, histone methylation, CpG islands, alphoid
repeats, gene orientation and gene deserts), failed to find
a statistical correlation between latency and genomic
features that were relevant for all five models of latency.
The features that affect latency seem to be largely local
and model specific76.
The problem of identifying replication-competent
viruses in patient-derived samples has hindered our
understanding of the relationship between HIV1 inte
gration sites and latency. Recent studies have shown
that almost all proviruses (98%) in resting CD4 +
Tcells from patients who are undergoing treatment
have major defects that preclude replication, such as
APOBEC3Ginduced hypermutation and large inter
nal deletions6,135. Latent proviruses that are replication-
PML competent are extremely rare and difficult to identify;
nuclear
body hence a detailed analysis of the factors that were pro
posed to contribute to latency in invitro models, such
as chromatin structure and histone modifications, could

Nature Reviews | Microbiology


NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 9

2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Promyelocytic leukaemia not really be studied invivo. However, recent studies of through additional rounds of activation. A minimal esti
bodies integration sites in patients who are undergoing pro mate of the inducible latent viral reservoir is provided
(PML bodies). Nuclear longed ART in whom clonally expanded HIV1infected by the viral outgrowth assay (VOA)6, whereas PCR-
structures the main component CD4 + T cells were identified, reinforced the role based approaches detect all proviruses and thus vastly
of which is the promyelocytic
leukaemia protein (or TRIM19).
of integration sites in the expansion and persistence of overestimate the size of the latent reservoir 138.
Several different protein HIV1 (REFS7,8082). Although the majority of viruses Although studies on material from patients infected
components as well as are defective, some clonally expanded cells contain with HIV represent the most valuable source of informa
various functions, such as replication-competent viruses. A study recently identi tion, an interesting insight into the correlation between
transcription, apoptosis,
fied a substantially expanded cellular clone in a patient integration sites and latency could also be obtained from
senescence DNA damage
response and antiviral defence,
with HIV1 and squamous cell carcinoma82. The clone experimental approaches. In this regard, the develop
are associated with these carried a replication-competent provirus that was inte ment of a dual colour (HIV Duo-Fluo I) virus system,
bodies. grated into a repetitive region of the host DNA; hence, which detects latently infected cells following infection,
the integration site could not be precisely localized. represents an interesting tool139,140.
CpG islands
Regions that have a high
Under current drug-treatment regimens, replication- Understanding the relevance of integration sites for
frequency of CpG sites, usually competent viruses persist within resting memory CD4+ the outcome of viral gene expression and infection may
connected with the promoter Tcells1,2,5 and can further be expanded through the have much broader implications. It might be relevant for
regions of genes. clonal amplification of these cells7,8082,136. An additional the development of gene therapy tools (vectors) that are
level of complexity was introduced by the finding that applicable in clinics and can have also important con
only a small proportion of the intact, non-defective pro notations for understanding the functions and networks
viruses in individuals undergoing ART were induced fol in the nucleus that are related to stem cell properties
lowing reactivation6. Out of 213 non-induced proviral andcancer.
clones, around 12% had intact genomes and were inte
grated into active transcriptional units, which suggests Therapeutic targeting of integration
that these replication-competent non-induced proviruses Antiretroviral drugs that are designed to target the
could become activated invivo. However, not all of these three essential viral enzymes, reverse transcriptase,
intact proviruses were induced to release virus follow protease and integrase, have a crucial role in ART. The
ing cellular activation in the standard viral outgrowth first ART regimens that were able to suppress viraemia
assay, which is often used to measure the latent reser to below the limit of detection in clinical assays con
voir 6. Repeated stimulation of infected cells from patients sisted of two nucleoside analogue reverse transcriptase
with HIV1 resulted in more viruses being reactivated inhibitors (NRTIs) in addition to a non-nucleoside
from latency than in the first round, which implies that reverse transcriptase inhibitor or an inhibitor of HIV1
reactivation from latency is a stochasticevent 6. protease141,142. Although regimens of this kind were the
The mechanisms that maintain HIV1 invivo were mainstay of ART for many years, they have been largely
recently explored in a detailed genetic analysis of viral replaced by regimens that consist of two NRTIs and an
sequences from individuals who have natural control integrase inhibitor 143 Thus, the discovery of HIV1 inte
of the infection (so-called HIV1 controllers). Marked grase inhibitors has had a major effect on treatment.
differences were identified between populations of A novel screen for compounds that blocked the strand
HIV1infected CD4+ Tcells in blood and lymphoid transfer reaction led to the discovery of this class of
tissues. Ongoing infection was reported to occur in drugs144. Raltegravir (RAL) is a prototypical integrase
follicular helper Tcells (TFH) and other memory CD4+ strand transfer inhibitor (INSTI). In combination with
subsets in lymph nodes. As some of these cells survive two NRTIs, RAL causes a very rapid decrease in viral
and return to the circulation, containing an inducible load145. The rapid decrease in viraemia results from
HIV1 genome, it was proposed that these cells link the fact that INSTIs act later in viral life cycle than
the pool of infected cells from lymphoid tissues to the other antiretrov iral drugs rather than from excep
pool of infected cells in blood. The latter pool of cells tional antiviral activity 146. INSTIs lack the cooperative
is represented mostly by highly differentiated, clon doseresponse curves that are responsible for the high
ally expanded CD4+ Tcells of the memory phenotype antiviral activity of the HIV1 protease inhibitors147.
that harbour archival viral sequences that usually are The clinical success of INSTIs results mainly from
refractory to activation137. favourable synergistic interactions with other antiretro
Proliferative self-renewal of the viral sequences in viral drugs and from the fact that they have few side
clonally expanded cells can indeed represent a selective effects148. The lack of side effects promotes adherence,
advantage for the provirus. However, it is unclear how which is crucial to the success of ART. If adherence is
clonal proliferation affects transcriptional processes and, suboptimal, patients develop resistance to RAL, indi
in particular, how it affects expression of the provirus. cating an urgent need to develop new generations of
These studies also point to the fact that integrated integrase inhibitors or find an alternative approach to
viruses in resting CD4+ Tcells are heterogeneous in target integration.
their nature and can indeed be divided into those that Elvitegravir (EVG) and dolutegravir (DTG) were
are induced to release replication-competent virus developed as a new generation of INSTIs. ELV shows
after one round of activation and those that are not some cross-resistance with RAL149. Resistance to DTG has
(non-induced). Most of the non-induced proviruses not yet proven to be a clinical problem, possibly because
are defective, but a portion is intact and can be induced of the high fitness cost of resistancemutations150,151.

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Cre recombinase Efforts have also been made to design inhibitors off-target effects, as well as technical challenges that are
A tyrosine recombinase that have a different mode of action than INSTIs. One related to their delivery to target cells, prevent their
enzyme that catalyses a such approach is to target integrase multimerization, clinical translation at this presenttime.
site-specific recombination which is a highly dynamic process of interactions The newest entry in the field of gene editing, the
event between two DNA
recognition sites; these sites
between different subunits. Several molecules that CRISPRCas9 system, has advantages over ZFNs and
consist of 13bp palindromic bind to theintegrase CCD dimer interface promote TALENs. DNA target specificity is determined by a 1723
sequences that flank an 8bp aberrantmultimerization152. nucleotide guide RNA that binds to Cas9 and directs it
spacer region. The unique and Another target is LEDGF interaction, which occurs to a complementary DNA for cleavage; the formed ribo
specific recombination system
through the IBD of LEDGF and the CCD of integrase153. nucleoprotein complex shows higher specificity than the
of the enzyme is used to
manipulate genes and
The proteinprotein interaction surface involves a proteinDNA complexes that are formed in ZFN and
chromosomes in several well-defined pocket, is limited in size and is composed TALEN systems and can be targeted to any sequence in
applications. of multiple hydrophobic interactions and hydrogen bond the genome. The system has already been used by differ
interactions; it thus represents an excellent starting point ent research groups to cleave and inactivate HIV1 DNA
Zinc-finger nucleases
(ZFNs). Artificial restriction
for the development of small-molecule inhibitors. The in human cells160163. These pioneering studies will possi
enzymes that are created first small-molecule quinoline-based inhibitors, which bly lead to future clinical applications with the purpose
through the fusion of a were reported to target the binding site in the CCD, were of either cleaving integrated latent HIV1 or inhibiting
zinc-finger DNA-binding termed integraseLEDGF inhibitors (LEDGINs)154 or viral replication once the problem of delivery issolved.
domain and a DNA-cleavage
allosteric integrase inhibitors (ALLINIs); they simul
domain. These enzymes
facilitate targeted editing of
taneously block the multimerization and allosterically Conclusions
thegenome by creating inhibit the enzymatic activity of integrase154. LEDGINs A huge effort has been made in the past few decades
double-strand breaks in the are not cross-resistant with INSTIs: the mode of action of towards a better understanding of the molecular mecha
DNA at a specific (desired) INSTIs requires the assembly of intasomes on the 3 ends nisms that govern the integration of HIV1 into the cellu
location.
of LTRs, whereas LEDGINs must be present before this lar genome. HIV1 preferentially targets transcriptionally
Transcription activator-like assembly to reach full efficacy. LEDGINs not only block active genes and open regions of chromatin. Inside the
effector nucleases integration but also decrease the replicative capacity of nuclear space not all active genes are targeted in the same
(TALENs). Restriction nucleases HIV1 by increasing the multimerization of integrase way, because HIV1 prefers active genes that are spatially
that are engineered to cut
during virion assembly 155. Efforts are being made to eval distributed in the outer shell of the nucleus. Cellular pro
specific DNA sequences.
uate LEDGINs for clinical use, but clinical trials have not teins that are involved in nuclear entry have implications
yet been carriedout. for integration that, although controlled by the viral inte
Another alternative approach is focused on targeting grase, depend on the chromatin-tethering factor LEDGF,
the integrated provirus rather than blocking the inte as well as on nuclear pore proteins. Despite the fact that
gration reaction. Strategies that are based on cleavage productive transcription usually follows integration,
of the integrated provirus have gained a lot of atten silencing of the viral genome can occur and persistent
tion in the past few years. These strategies are based on viral reservoirs, which are a major hurdle in curing infec
programmable nucleases that specifically bind to, and tion with HIV1, are formed. Very intriguing questions
cleave, integrated HIV1 DNA. Among the first enzy that still need to be answered are to what extent are inte
matic candidates is a Cre recombinase (Tre) that binds to, gration and viral transcription connected and how do sites
and cuts, the LTR156,157; more recently, engineered zinc- of integration define the transcriptional activity of the
finger nucleases (ZFNs) or transcription activator-like effector provirus. Most importantly, is there a direct connection
nucleases (TALENs) were developed as promising new between integration and silencing of the viral genome?
tools for nuclease-based HIV1 therapy. These nucle Although there are indications that HIV integration might
ases were designed with the goal of decreasing off-target be tightly connected with the establishment and mainte
mutations and obtaining higher nuclease activity 158,159. nance of latency this requires further and more definitive
Despite the great potential of nuclease-based approaches, confirmation, and will be a subject of future studies.

1. Chun,T.W. etal. Quantification of latent tissue patients are non-inducible because they are 11. Yamashita,M. & Emerman,M. Capsid is a
reservoirs and total body viral load in HIV1 infection. highlydefective. A portion of these non-induced dominantdeterminant of retrovirus infectivity in
Nature 387, 183188 (1997). proviruses have intact genomes and can be induced nondividing cells. J.Virol. 78, 56705678 (2004).
2. Finzi,D. etal. Identification of a reservoir for HIV1 after several rounds of stimulation. 12. Yamashita,M., Perez,O., Hope,T.J. & Emerman,M.
inpatients on highly active antiretroviral therapy. 7. Cohn,L.B. etal. HIV1 integration landscape Evidence for direct involvement of the capsid protein
Science 278, 12951300 (1997). duringlatent and active infection. Cell 160, 420432 in HIV infection of nondividing cells. PLoS Pathog. 3,
3. Wong,J.K. etal. Recovery of replication-competent (2015). 15021510 (2007).
HIV despite prolonged suppression of plasma viremia. Together with references 80 and 81, this work 13. Lee,K. etal. Flexible use of nuclear import pathways
Science 278, 12911295 (1997). shows that HIV1 integrates into genes that are by HIV1. Cell Host Microbe 7, 221233 (2010).
4. Chun,T.W. etal. Presence of an inducible HIV1 associated with cellular proliferation and clonal A study that identifies truncated CPSF6 as a
latentreservoir during highly active antiretroviral expansion. It also suggests that most of the dominant negative inhibitor of HIV1 infection
therapy. Proc. Natl Acad. Sci. USA 94, 1319313197 proviruses in dividing clonally expanded Tcells and in which CPSP6 is shown to have a role in
(1997). aredefective. targeting HIV1 to use specific cofactors.
5. Finzi,D. etal. Latent infection of CD4+ Tcells provides 8. Coffin,J., Hughes,S. & Varmus,H. (eds) Retroviruses. 14. Schaller,T. etal. HIV1 capsid-cyclophilin
a mechanism for lifelong persistence of HIV1, even in (Cold Spring Harbor Laboratory Press, 1997). interactions determine nuclear import pathway,
patients on effective combination therapy. Nat. Med. 9. Suzuki,Y. & Craigie,R. The road to chromatin integration targeting and replication efficiency. PLoS
5, 512517 (1999). nuclear entry of retroviruses. Nat. Rev. Microbiol. 5, Pathog. 7, e1002439 (2011).
6. Ho,Y.C. etal. Replication-competent noninduced 187196 (2007). This work connects, for the first time, the capsid of
proviruses in the latent reservoir increase barrier 10. Matreyek,K.A. & Engelman,A. Viral and cellular HIV1 with the selection of integration site by
toHIV1 cure. Cell 155, 540551(2013). requirements for the nuclear entry of retroviral showing that capsid mutants that do not recruit
This study demonstrates that the majority of preintegration nucleoprotein complexes. Viruses 5, CPSF6 or CYPA integrate into different target
proviruses in resting CD4+ T cells from treated 24832511 (2013). regions of the genome.

NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 11



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

15. Peng,K. etal. Quantitative microscopy of functional HIV integration to transcriptionally active chromatin. 60. Lewinski,M.K. etal. Retroviral DNA integration: viral
post-entry complexes reveals association of replication Proc.Natl Acad. Sci. USA 113, E1054E1063 and cellular determinants of target-site selection.
with the viral capsid. eLife 3, e04114 (2014). (2016). PLoSPathog. 2, e60 (2006).
16. Koh,Y. etal. Differential effects of human This work shows, for the first time, that CPSF6, as a 61. Krishnan,L. etal. Structure-based modeling of the
immunodeficiency virus type1 capsid and cellular capsid-binding protein, has a role in directing HIV1 functional HIV1 intasome and its inhibition. Proc. Natl
factors nucleoporin 153 and LEDGF/p75 on the integration to transcriptionally active chromatin Acad. Sci. USA 107, 1591015915 (2010).
efficiency and specificity of viral DNA integration. regions, whereas LEDGF, as an integrase partner, 62. Vink,C. etal. Analysis of the junctions between human
J.Virol. 87, 648658 (2013). directs integration into genes. immunodeficiency virus type1 proviral DNA and
17. Bukrinsky,M.I. etal. A nuclear localization signal 41. Arhel,N.J. etal. HIV1 DNA flap formation promotes human DNA. J.Virol. 64, 56265627 (1990).
within HIV1 matrix protein that governs infection of uncoating of the pre-integration complex at the 63. Muller,H.P. & Varmus,H.E. DNA bending creates
non-dividing cells. Nature 365, 666669 (1993). nuclear pore. EMBO J. 26, 30253037 (2007). favored sites for retroviral integration: an explanation
18. Reil,H., Bukovsky,A.A., Gelderblom,H.R. 42. Craigie,R. & Bushman,F.D. HIV DNA integration. for preferred insertion sites in nucleosomes. EMBO J.
&Gottlinger,H.G. Efficient HIV1 replication can occur Cold Spring Harb. Perspect. Med. 2, a006890 13, 47044714 (1994).
in the absence of the viral matrix protein. EMBO J. 17, (2012). 64. Pryciak,P.M. & Varmus,H.E. Nucleosomes, DNA-
26992708 (1998). 43. Craigie,R. & Bushman,F.D. Host factors in retroviral binding proteins, and DNA sequence modulate
19. Guenzel,C.A., Herate,C. & Benichou,S. HIV1 Vpr integration and the selection of integration target retroviral integration target site selection. Cell 69,
astill enigmatic multitasker. Front. Microbiol. 5, 127 sites. Microbiol. Spectr. 2, MDNA300262014 769780 (1992).
(2014). (2014). 65. Serrao,E. etal. Integrase residues that determine
20. Popov,S. etal. Viral protein R regulates nuclear import 44. Engelman,A., Bushman,F.D. & Craigie,R. nucleotide preferences at sites of HIV1 integration:
of the HIV1 pre-integration complex. EMBO J. 17, Identification of discrete functional domains of HIV1 implications for the mechanism of target DNA binding.
909917 (1998). integrase and their organization within an active Nucleic Acids Res. 42, 51645176 (2014).
21. Fouchier,R.A. etal. Interaction of the human multimeric complex. EMBO J. 12, 32693275 66. Demeulemeester,J. etal. HIV1 integrase variants
immunodeficiency virus type1Vpr protein with the (1993). retarget viral integration and are associated with
nuclear pore complex. J.Virol. 72, 60046013 (1998). 45. van Gent,D.C., Vink,C., Groeneger,A.A. disease progression in a chronic infection cohort.
22. McDonald,D. etal. Visualization of the intracellular &Plasterk,R.H. Complementation between HIV CellHost Microbe 16, 651662 (2014).
behavior of HIV in living cells. J.Cell Biol. 159, integrase proteins mutated in different domains. 67. Holman,A.G. & Coffin,J.M. Symmetrical base
441452 (2002). EMBO J. 12, 32613267 (1993). preferences surrounding HIV1, avian sarcoma/leukosis
23. Brass,A.L. etal. Identification of host proteins required 46. Dyda,F. etal. Crystal structure of the catalytic domain virus, and murine leukemia virus integration sites.
for HIV infection through a functional genomic screen. of HIV1 integrase: similarity to other polynucleotidyl Proc. Natl Acad. Sci. USA 102, 61036107 (2005).
Science 319, 921926 (2008). transferases. Science 266, 19811986 (1994). 68. Brady,T. etal. HIV integration site distributions in
24. Konig,R. etal. Global analysis of hostpathogen 47. Craigie,R. HIV integrase, a brief overview from resting and activated CD4+ Tcells infected in culture.
interactions that regulate early-stage HIV1 replication. chemistry to therapeutics. J.Biol. Chem. 276, AIDS 23, 14611471 (2009).
Cell 135, 4960 (2008). 2321323216 (2001). 69. Maskell,D.P. etal. Structural basis for retroviral
Together with reference 23, this work describes a 48. Carayon,K. etal. A cooperative and specific DNA- integration into nucleosomes. Nature 523, 366369
high-throughput screening that identifies several binding mode of HIV1 integrase depends on the (2015).
cellular factors that are involved in HIV1 infection. nature of the metallic cofactor and involves the zinc- This work shows, through the use of single-particle
25. Bushman,F.D. etal. Host cell factors in HIV containing Nterminal domain. Nucleic Acids Res. 38, cryoEM, how the PFV intasome as a viral DNA
replication: meta-analysis of genome-wide studies. 36923708 (2010). recombination machinery captures nucleosomes to
PLoS Pathog. 5, e1000437 (2009). 49. Kulkosky,J., Jones,K.S., Katz,R.A., Mack,J.P. enable integration.
26. Di Nunzio,F. etal. Nup153 and Nup98 bind the HIV1 &Skalka,A.M. Residues critical for retroviral 70. Demeulemeester,J., De Rijck,J., Gijsbers,R.
core and contribute to the early steps of HIV1 integrative recombination in a region that is highly &Debyser,Z. Retroviral integration: site matters:
replication. Virology 440, 818 (2013). conserved among retroviral/retrotransposon mechanisms and consequences of retroviral
27. Matreyek,K.A., Yucel,S.S., Li,X. & Engelman,A. integrases and bacterial insertion sequence integration site selection. Bioessays 37, 12021214
Nucleoporin NUP153 phenylalanine-glycine motifs transposases. Mol. Cell. Biol. 12, 23312338 (2015).
engage a common binding pocket within the HIV1 (1992). 71. Bushman,F. etal. Genome-wide analysis of retroviral
capsid protein to mediate lentiviral infectivity. 50. Leavitt,A.D., Shiue,L. & Varmus,H.E. Site-directed DNA integration. Nat. Rev. Microbiol. 3, 848858
PLoSPathog. 9, e1003693 (2013). mutagenesis of HIV1 integrase demonstrates (2005).
28. Gamble,T.R. etal. Crystal structure of human differential effects on integrase functions invitro. 72. Schroder,A.R. etal. HIV1 integration in the human
cyclophilin A bound to the amino-terminal domain of J.Biol. Chem. 268, 21132119 (1993). genome favors active genes and local hotspots. Cell
HIV1 capsid. Cell 87, 12851294 (1996). 51. Manganaro,L. etal. Concerted action of cellular JNK 110, 521529 (2002).
29. Ocwieja,K.E. etal. HIV integration targeting: and Pin1 restricts HIV1 genome integration to The first study to define HIV1 integration sites
apathway involving Transportin3 and the nuclear pore activated CD4+ T lymphocytes. Nat. Med. 16, genome-wide through the use of whole-genome
protein RanBP2. PLoS Pathog. 7, e1001313 (2011). 329333 (2010). sequencing. This study shows that HIV1 favours
30. Bosco,D.A., Eisenmesser,E.Z., Pochapsky,S., 52. Lutzke,R.A., Vink,C. & Plasterk,R.H. integration into active transcriptional units.
Sundquist,W.I. & Kern,D. Catalysis of cis/trans Characterization of the minimal DNA-binding domain 73. Mitchell,R.S. etal. Retroviral DNA integration: ASLV,
isomerization in native HIV1 capsid by human of the HIV integrase protein. Nucleic Acids Res. 22, HIV, and MLV show distinct target site preferences.
cyclophilin A. Proc. Natl Acad. Sci. USA 99, 41254131 (1994). PLoS Biol. 2, E234 (2004).
52475252 (2002). 53. Eijkelenboom,A.P. etal. The solution structure of the 74. Han,Y. etal. Resting CD4+ Tcells from human
31. Christ,F. etal. TransportinSR2 imports HIV into the amino-terminal HHCC domain of HIV2 integrase: a immunodeficiency virus type1 (HIV1)-infected
nucleus. Curr. Biol. 18, 11921202 (2008). three-helix bundle stabilized by zinc. Curr. Biol. 7, individuals carry integrated HIV1 genomes within
32. Kataoka,N., Bachorik,J.L. & Dreyfuss,G. 739746 (1997). actively transcribed host genes. J.Virol. 78,
TransportinSR, a nuclear import receptor for SR 54. Cereseto,A. etal. Acetylation of HIV1 integrase by 61226133 (2004).
proteins. J.Cell Biol. 145, 11451152 (1999). p300 regulates viral integration. EMBO J. 24, 75. Barr,S.D. etal. HIV integration site selection:
33. Lee,K. etal. HIV1 capsid-targeting domain of cleavage 30703081 (2005). targeting in macrophages and the effects of different
and polyadenylation specificity factor 6. J.Virol. 86, 55. Savarino,A. In silico docking of HIV1 integrase routes of viral entry. Mol. Ther. 14, 218225
38513860 (2012). inhibitors reveals a novel drug type acting on an (2006).
34. Bhattacharya,A. etal. Structural basis of HIV1 capsid enzyme/DNA reaction intermediate. Retrovirology 4, 76. Sherrill-Mix,S. etal. HIV latency and integration site
recognition by PF74 and CPSF6. Proc. Natl Acad. 21 (2007). placement in five cell-based models. Retrovirology
Sci.USA 111, 1862518630 (2014). 56. Hare,S., Gupta,S.S., Valkov,E., Engelman,A. 10, 90 (2013).
35. Rasheedi,S. etal. The cleavage and polyadenylation &Cherepanov,P. Retroviral intasome assembly and 77. Wang,G.P., Ciuffi,A., Leipzig,J., Berry,C.C.
specificity factor 6 (CPSF6) subunit of the capsid- inhibition of DNA strand transfer. Nature 464, &Bushman,F.D. HIV integration site selection:
recruited pre-messenger RNA cleavage factor I (CFIm) 232236 (2010). analysis by massively parallel pyrosequencing
complex mediates HIV1 integration into genes. Together with reference 57, this study resolves the revealsassociation with epigenetic modifications.
J.Biol.Chem. 291, 1180911819 (2016). Xray crystal structure of the PFV intasome, which Genome Res. 17, 11861194 (2007).
36. Chin,C.R. etal. Direct visualization of HIV1 has substantially contributed to our understanding 78. Ferris,A.L. etal. Lens epithelium-derived
replication intermediates shows that capsid and of the mechanisms of retroviral-DNA integration. growthfactor fusion proteins redirect HIV1
CPSF6 modulate HIV1 intra-nuclear invasion and 57. Maertens,G.N., Hare,S. & Cherepanov,P. The DNA integration. Proc. Natl Acad. Sci. USA 107,
integration. Cell Rep. 13, 17171731 (2015). mechanism of retroviral integration from Xray 31353140 (2010).
37. Ruegsegger,U., Blank,D. & Keller,W. Human structures of its key intermediates. Nature 468, 79. Singh,P.K. etal. LEDGF/p75 interacts with mRNA
pre-mRNA cleavage factor Im is related to 326329 (2010). splicing factors and targets HIV1 integration to highly
spliceosomal SRproteins and can be reconstituted Similar to reference 56, this study shows the spliced genes. Genes Dev. 29, 22872297 (2015).
invitro from recombinant subunits. Mol. Cell 1, Xraycrystal structure of the PFV intasome in 80. Wagner,T.A. etal. HIV latency. Proliferation of cells
243253 (1998). complex with target DNA before, and following, with HIV integrated into cancer genes contributes to
38. De Iaco,A. etal. TNPO3 protects HIV1 replication strand transfer. persistent infection. Science 345, 570573 (2014).
from CPSF6mediated capsid stabilization in the host 58. Bushman,F.D., Fujiwara,T. & Craigie,R. Retroviral 81. Maldarelli,F. etal. HIV latency. Specific HIV integration
cell cytoplasm. Retrovirology 10, 20 (2013). DNA integration directed by HIV integration protein sites are linked to clonal expansion and persistence of
39. Hilditch,L. & Towers,G.J. A model for cofactor use invitro. Science 249, 15551558 (1990). infected cells. Science 345, 179183 (2014).
during HIV1 reverse transcription and nuclear entry. 59. Craigie,R., Fujiwara,T. & Bushman,F. The IN protein References 80 and 81 connect HIV1 integration
Curr. Opin. Virol. 4, 3236 (2014). of Moloney murine leukemia virus processes the viral with the clonal expansion of target cells and propose
40. Sowd,G.A. etal. A critical role for alternative DNA ends and accomplishes their integration invitro. that this could contribute to the aberrant expansion
polyadenylation factor CPSF6 in targeting HIV1 Cell 62, 829837 (1990). of viral genomes and latent reservoirs.

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

82. Simonetti,F.R. etal. Clonally expanded CD4+ Tcells 105. Lesbats,P. etal. Functional coupling between HIV1 132. Dieudonne,M. etal. Transcriptional competence of
can produce infectious HIV1 invivo. Proc. Natl Acad. integrase and the SWI/SNF chromatin remodeling the integrated HIV1 provirus at the nuclear periphery.
Sci. USA 113, 18831888 (2016). complex for efficient invitro integration into stable EMBO J. 28, 22312243 (2009).
This work provides evidence that clonally expanded nucleosomes. PLoS Pathog. 7, e1001280 (2011). 133. Lusic,M. etal. Proximity to PML nuclear bodies
HIV1infected cells contain replication-competent 106. Allouch,A. etal. The TRIM family protein KAP1 inhibits regulates HIV1 latency in CD4+ Tcells. Cell Host
viruses. HIV1 integration. Cell Host Microbe 9, 484495 Microbe 13, 665677 (2013).
83. Ikeda,T., Shibata,J., Yoshimura,K., Koito,A. (2011). 134. Jordan,A., Bisgrove,D. & Verdin,E. HIV reproducibly
&Matsushita,S. Recurrent HIV1 integration at the 107. Quercioli,V. etal. Comparative analysis of HIV1 and establishes a latent infection after acute infection of
BACH2 locus in resting CD4+ Tcell populations during murine leukemia virus three-dimensional nuclear Tcells invitro. EMBO J. 22, 18681877 (2003).
effective highly active antiretroviral therapy. J.Infect. Dis. distributions. J.Virol. 90, 52055209 (2016). This work describes the generation of latently
195, 716725 (2007). 108. Cremer,T. etal. Chromosome territories a functional infected Jurkat cells (JLats), which, to date, remain
84. Kobayashi,S. etal. Identification of IGHCBACH2 nuclear landscape. Curr. Opin. Cell Biol. 18, 307316 one of the most widely used clonal models of
fusion transcripts resulting from cryptic chromosomal (2006). latency. Integration sites were proposed to
rearrangements of 14q32 with 6q15 in aggressive 109. Cavalli,G. & Misteli,T. Functional implications of genome correlate with the silencing of the viral genome.
Bcell lymphoma/leukemia. Genes Chromosomes topology. Nat. Struct. Mol. Biol. 20, 290299 (2013). 135. Bruner,K.M. etal. Defective proviruses rapidly
Cancer 50, 207216 (2011). 110. Burdick,R.C., Hu,W.S. & Pathak,V.K. Nuclear import accumulate during acute HIV1 infection. Nat. Med.
85. Flucke,U. etal. Presence of C11orf95MKL2 fusion is of APOBEC3Flabeled HIV1 preintegration complexes. 22, 10431049 (2016).
a consistent finding in chondroid lipomas: a study of Proc. Natl Acad. Sci. USA 110, E4780E4789 (2013). 136. Kim,M. & Siliciano,R.F. Reservoir expansion by Tcell
eight cases. Histopathology 62, 925930 (2013). 111. Guelen,L. etal. Domain organization of human proliferation may be another barrier to curing HIV
86. Liu,H. etal. Integration of human immunodeficiency chromosomes revealed by mapping of nuclear lamina infection. Proc. Natl Acad. Sci. USA 113, 16921694
virus type1 in untreated infection occurs preferentially interactions. Nature 453, 948951 (2008). (2016).
within genes. J.Virol. 80, 77657768 (2006). 112. Ibarra,A. & Hetzer,M.W. Nuclear pore proteins and 137. Boritz,E.A. etal. Multiple origins of virus persistence
87. Marini,B. etal. Nuclear architecture dictates HIV1 the control of genome functions. Genes Dev. 29, during natural control of HIV infection. Cell 166,
integration site selection. Nature 521, 227231 337349 (2015). 10041015 (2016).
(2015). 113. Raices,M. & DAngelo,M.A. Nuclear pore complex This study shows that, in individuals that have
This work shows that HIV1 positions its genome in composition: a new regulator of tissue-specific and natural control of HIV1 replication, three
regions in proximity to the NPC at the nuclear developmental functions. Nat. Rev. Mol. Cell Biol. 13, mechanisms contribute to HIV replication in
periphery. 687699 (2012). anatomically and functionally distinct compartments.
88. Albanese,A., Arosio,D., Terreni,M. & Cereseto,A. 114. Lelek,M. etal. Chromatin organization at the nuclear 138. Eriksson,S. etal. Comparative analysis of measures
HIV1 pre-integration complexes selectively target pore favours HIV replication. Nat. Commun. 6, 6483 of viral reservoirs in HIV1 eradication studies.
decondensed chromatin in the nuclear periphery. PloS (2015). PLoSPathog. 9, e1003174 (2013).
ONE 3, e2413 (2008). 115. Krull,S. etal. Protein Tpr is required for establishing 139. Calvanese,V., Chavez,L., Laurent,T., Ding,S. &
89. Wu,X., Li,Y., Crise,B. & Burgess,S.M. Transcription nuclear pore-associated zones of heterochromatin Verdin,E. Dual-color HIV reporters trace a population
start regions in the human genome are favored exclusion. EMBO J. 29, 16591673 (2010). of latently infected cells and enable their purification.
targetsfor MLV integration. Science 300, 17491751 116. Wong,R.W., Mamede,J.I. & Hope,T.J. Impact of Virology 446, 283292 (2013).
(2003). nucleoporin-mediated chromatin localization and 140. Dahabieh,M.S., Ooms,M., Simon,V. & Sadowski,I.
90. Ge,H., Si,Y. & Roeder,R.G. Isolation of cDNAs nuclear architecture on HIV integration site selection. A doubly fluorescent HIV1 reporter shows that the
encoding novel transcription coactivators p52 and p75 J.Virol. 89, 97029705 (2015). majority of integrated HIV1 is latent shortly after
reveals an alternate regulatory mechanism of 117. Van Lint,C., Bouchat,S. & Marcello,A. HIV1 infection. J.Virol. 87, 47164727 (2013).
transcriptional activation. EMBO J. 17, 67236729 transcription and latency: an update. Retrovirology 10, 141. Gulick,R.M. etal. Treatment with indinavir,
(1998). 67 (2013). zidovudine, and lamivudine in adults with human
91. Singh,D.P. etal. Lens epithelium-derived growth 118. Lusic,M. & Giacca,M. Regulation of HIV1 latency immunodeficiency virus infection and prior
factor: effects on growth and survival of lens epithelial bychromatin structure and nuclear architecture. antiretroviral therapy. N. Engl. J.Med. 337,
cells, keratinocytes, and fibroblasts. Biochem. Biophys. J.Mol.Biol. 427, 688694 (2014). 734739 (1997).
Res. Commun. 267, 373381 (2000). 119. Dahabieh,M.S., Battivelli,E. & Verdin,E. 142. Perelson,A.S. etal. Decay characteristics of
92. Cherepanov,P. etal. HIV1 integrase forms stable Understanding HIV latency: the road to an HIV cure. HIV1infected compartments during combination
tetramers and associates with LEDGF/p75 protein in Annu. Rev. Med. 66, 407421 (2015). therapy. Nature 387, 188191 (1997).
human cells. J.Biol. Chem. 278, 372381 (2003). 120. du Chene,I. etal. Suv39H1 and HP1 are responsible 143. Gunthard,H.F. etal. Antiretroviral drugs for
93. Ciuffi,A. etal. A role for LEDGF/p75 in targeting for chromatin-mediated HIV1 transcriptional silencing treatment and prevention of HIV infection in adults:
HIVDNA integration. Nat. Med. 11, 12871289 and post-integration latency. EMBO J. 26, 424435 2016 recommendations of the International
(2005). (2007). Antiviral SocietyUSA Panel. JAMA 316, 191210
94. Llano,M. etal. An essential role for LEDGF/p75 in HIV 121. Imai,K., Togami,H. & Okamoto,T. Involvement of (2016).
integration. Science 314, 461464 (2006). histone H3 lysine 9 (H3K9) methyltransferase G9a in 144. Hazuda,D.J. etal. Inhibitors of strand transfer that
95. Maertens,G. etal. LEDGF/p75 is essential for nuclear the maintenance of HIV1 latency and its reactivation prevent integration and inhibit HIV1 replication in
and chromosomal targeting of HIV1 integrase in by BIX01294. J.Biol. Chem. 285, 1653816545 cells. Science 287, 646650 (2000).
human cells. J.Biol. Chem. 278, 3352833539 (2003). (2010). One of the first screenings of chemical compounds
96. Schrijvers,R. etal. LEDGF/p75independent HIV1 122. Friedman,J. etal. Epigenetic silencing of HIV1 by the that block the strand transfer activity of
replication demonstrates a role for HRP2 and remains histone H3 lysine 27 methyltransferase enhancer of HIV1integrase, which led to the identification of
sensitive to inhibition by LEDGINs. PLoS Pathog. 8, Zeste 2. J.Virol. 85, 90789089 (2011). raltegravir.
e1002558 (2012). 123. Kauder,S.E., Bosque,A., Lindqvist,A., Planelles,V. 145. Christ,F. & Debyser,Z. HIV1 integrase inhibition:
97. Shun,M.C. etal. LEDGF/p75 functions downstream &Verdin,E. Epigenetic regulation of HIV1 latency by looking at cofactor interactions. Future Med. Chem.
from preintegration complex formation to effect gene- cytosine methylation. PLoS Pathog. 5, e1000495 7, 24072410 (2015).
specific HIV1 integration. Genes Dev. 21, 17671778 (2009). 146. Sedaghat,A.R., Dinoso,J.B., Shen,L., Wilke,C.O.
(2007). 124. Blazkova,J. etal. CpG methylation controls & Siliciano,R.F. Decay dynamics of HIV1 depend on
98. Debyser,Z., Christ,F., De Rijck,J. & Gijsbers,R. reactivation of HIV from latency. PLoS Pathog. 5, the inhibited stages of the viral life cycle. Proc. Natl
Hostfactors for retroviral integration site selection. e1000554 (2009). Acad. Sci. USA 105, 48324837 (2008).
Trends Biochem. Sci. 40, 108116 (2015). 125. Marcello,A. etal. Recruitment of human cyclin T1 to 147. Shen,L. etal. Dose-response curve slope sets class-
99. Pradeepa,M.M., Sutherland,H.G., Ule,J., nuclear bodies through direct interaction with the PML specific limits on inhibitory potential of anti-HIV
Grimes,G.R. & Bickmore,W.A. Psip1/Ledgf p52 binds protein. EMBO J. 22, 21562166 (2003). drugs. Nat. Med. 14, 762766 (2008).
methylated histone H3K36 and splicing factors and 126. Sabo,A., Lusic,M., Cereseto,A. & Giacca,M. 148. Jilek,B.L. etal. A quantitative basis for antiretroviral
contributes to the regulation of alternative splicing. Acetylation of conserved lysines in the catalytic core of therapy for HIV1 infection. Nat. Med. 18, 446451
PLoS Genet. 8, e1002717 (2012). cyclin-dependent kinase 9 inhibits kinase activity and (2012).
100. Eidahl,J.O. etal. Structural basis for high-affinity regulates transcription. Mol. Cell. Biol. 28, 149. Zolopa,A.R. etal. Activity of elvitegravir, a once-
binding of LEDGF PWWP to mononucleosomes. 22012212 (2008). daily integrase inhibitor, against resistant HIV type1:
NucleicAcids Res. 41, 39243936 (2013). 127. Williams,S.A., Kwon,H., Chen,L.F. & Greene,W.C. results of a phase2, randomized, controlled, dose-
101. Van Steensel,B. & Henikoff,S. Identification of invivo Sustained induction of NFB is required for efficient ranging clinical trial. J.Infect. Dis. 201, 814822
DNA targets of chromatin proteins using tethered dam expression of latent human immunodeficiency virus (2010).
methyltransferase. Nat. Biotechnol. 18, 424428 type1. J.Virol. 81, 60436056 (2007). 150. Garrido,C. etal. Resistance associated mutations to
(2000). 128. Lenasi,T., Contreras,X. & Peterlin,B.M. dolutegravir (S/GSK1349572) in HIV-infected patients
102. De Rijck,J., Bartholomeeusen,K., Ceulemans,H., Transcriptional interference antagonizes proviral gene impact of HIV subtypes and prior raltegravir
Debyser,Z. & Gijsbers,R. High-resolution profiling of expression to promote HIV latency. Cell Host Microbe experience. Antiviral Res. 90, 164167 (2011).
the LEDGF/p75 chromatin interaction in the ENCODE 4, 123133 (2008). 151. Brenner,B.G. & Wainberg,M.A. Clinical benefit of
region. Nucleic Acids Res. 38, 61356147 (2010). 129. Han,Y. etal. Orientation-dependent regulation of dolutegravir in HIV1 management related to the high
103. Gijsbers,R. etal. Role of the PWWP domain of lens integrated HIV1 expression by host gene genetic barrier to drug resistance. Virus Res. http://
epithelium-derived growth factor (LEDGF)/p75 cofactor transcriptional readthrough. Cell Host Microbe 4, dx.doi.org/10.1016/j.virusres.2016.07.006 (2016).
in lentiviral integration targeting. J.Biol. Chem. 286, 134146 (2008). 152. Kessl,J.J. etal. An allosteric mechanism for inhibiting
4181241825 (2011). 130. Shan,L. etal. Influence of host gene transcription level HIV1 integrase with a small molecule. Mol.
104. Farnet,C.M. & Bushman,F.D. HIV1 cDNA and orientation on HIV1 latency in a primary-cell Pharmacol. 76, 824832 (2009).
integration: requirement of HMG I(Y) protein for model. J.Virol. 85, 53845393 (2011). 153. Cherepanov,P. etal. Solution structure of the HIV1
function of preintegration complexes invitro. Cell 88, 131. Siliciano,R.F. & Greene,W.C. HIV latency. Cold Spring integrase-binding domain in LEDGF/p75. Nat. Struct.
483492 (1997). Harb. Perspect. Med. 1, a007096 (2011). Mol. Biol. 12, 526532 (2005).

NATURE REVIEWS | MICROBIOLOGY ADVANCE ONLINE PUBLICATION | 13



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

154. Christ,F. etal. Rational design of small-molecule 160. Ebina,H., Misawa,N., Kanemura,Y. & Koyanagi,Y. References 164166 are comprehensive recent
inhibitors of the LEDGF/p75integrase interaction Harnessing the CRISPR/Cas9 system to disrupt latent reviews of the genomic regulatory landscapes and
and HIV replication. Nat. Chem. Biol. 6, 442448 HIV1 provirus. Sci. Rep. 3, 2510 (2013). nuclear architecture.
(2010). 161. Hu,W. etal. RNA-directed gene editing specifically 167. Dundr,M. Nuclear bodies: multifunctional companions
This study details a structure-based rational design eradicates latent and prevents new HIV1 infection. of the genome. Curr. Opin. Cell Biol. 24, 415422
of LEDGINs, integrase and LEDGF interaction Proc. Natl Acad. Sci. USA 111, 1146111466 (2012).
inhibitors with antiviral activity. (2014). 168. Kind,J. etal. Single-cell dynamics of genome-nuclear
155. Desimmie,B.A. etal. LEDGINs inhibit late stage HIV1 162. Liao,H.K. etal. Use of the CRISPR/Cas9 system as an lamina interactions. Cell 153, 178192 (2013).
replication by modulating integrase multimerization in intracellular defense against HIV1 infection in human 169. Harr,J.C. etal. Directed targeting of chromatin to
the virions. Retrovirology 10, 57 (2013). cells. Nat. Commun. 6, 6413 (2015). the nuclear lamina is mediated by chromatin state
156. Sarkar,I., Hauber,I., Hauber,J. & Buchholz,F. HIV1 163. Wang,G., Zhao,N., Berkhout,B. & Das,A.T. and Atype lamins. J.Cell Biol. 208, 3352 (2015).
proviral DNA excision using an evolved recombinase. CRISPRCas9 can inhibit HIV1 replication but 170. Schermelleh,L. etal. Subdiffraction multicolor
Science 316, 19121915 (2007). NHEJ repair facilitates virus escape. Mol. Ther. 24, imaging of the nuclear periphery with 3D structured
157. Hauber,I. etal. Highly significant antiviral activity of 522526 (2016). illumination microscopy. Science 320, 13321336
HIV1 LTR-specific tre-recombinase in humanized mice. 164. Dekker,J. & Misteli,T. Long-range chromatin (2008).
PLoS Pathog. 9, e1003587 (2013). interactions. Cold Spring Harb. Perspect. Biol. 7,
158. Qu,X. etal. Zinc-finger-nucleases mediate specific and a019356 (2015). Acknowledgements
efficient excision of HIV1 proviral DNA from infected 165. Dixon,J.R., Gorkin,D.U. & Ren,B. Chromatin M.L. is funded by grants from the German Centre for Infectious
and latently infected human Tcells. Nucleic Acids Res. domains: the unit of chromosome organization. Research (Deutsche Zentrum fur Infektion Forschung, DZIF)
41, 77717782 (2013). Mol.Cell 62, 668680 (2016). and by the Hector Foundation for AIDS and Cancer Research.
159. Strong,C.L. etal. Damaging the integrated HIV 166. Bonev,J. & Cavaalli,G. Organization and function of
proviral DNA with TALENs. PloS ONE 10, e0125652 the 3D genome. Nat. Rev. Genet. 17, 661678 Competing interests statement
(2015). (2016). The authors declare no competing interests.

14 | ADVANCE ONLINE PUBLICATION www.nature.com/nrmicro



2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like