SUMMARY
The HIV-1 capsid, composed of approximately 1,200 copies of the capsid protein, encases genomic RNA alongside viral nucleocapsid, reverse transcriptase, and integrase proteins. After cell entry, the capsid interacts with a myriad of host factors to traverse the cell cytoplasm, pass through the nuclear pore complex (NPC), and then traffic to chromosomal sites for viral DNA integration. Integration may very well require the dissolution of the capsid, but where and when this uncoating event occurs remains hotly debated. Based on size constraints, a long-prevailing view was that uncoating preceded nuclear transport, but recent research has indicated that the capsid may remain largely intact during nuclear import, with perhaps some structural remodeling required for NPC traversal. Completion of reverse transcription in the nucleus may further aid capsid uncoating. One canonical type of host factor, typified by CPSF6, leverages a Phe-Gly (FG) motif to bind capsid. Recent research has shown these peptides reside amid prion-like domains (PrLDs), which are stretches of protein sequence devoid of charged residues. Intermolecular PrLD interactions along the exterior of the capsid shell impart avid host factor binding for productive HIV-1 infection. Herein we overview capsid–host interactions implicated in HIV-1 ingress and discuss important research questions moving forward. Highlighting clinical relevance, the long-acting ultrapotent inhibitor lenacapavir, which engages the same capsid binding pocket as FG host factors, was recently approved to treat people living with HIV.
KEYWORDS: HIV, capsid, trafficking, nuclear import, CPSF6, NUP153, FG domain, prion-like domain, mixed-charge domain, liquid-liquid phase separation, speckle-associated domain, lenacapavir
INTRODUCTION
HIV/AIDS is a significant global health problem. At the same time, antiretroviral therapy (ART) has made a historical impact on the course of the HIV-1 pandemic. Prior to ART, HIV-1 infection in the vast majority of cases led to acquired immunodeficiency syndrome and death. Current ART regimens are typically composed of mixtures of antiviral compounds that target critical viral enzyme activities, including reverse transcriptase (RT) and integrase activities. Despite the huge positive impact of ART on public health, drug resistance is not uncommon, necessitating the development of new compounds that are active against drug resistant HIV-1 strains as well as new classes of drugs that target previously unexploited viral targets. Lenacapavir, which is a first-in-class capsid inhibitor, was recently approved to treat HIV-1 infection (1).
The capsid is a key substructure of the HIV-1 virus particle. HIV-1 particles assemble at the plasma membrane, through which the fledgling viruses bud (2). The resulting lipid-enveloped virions are roughly spherical, ~90–120 nm in diameter. Viral structural proteins [matrix (MA), nucleocapsid (NC), and capsid protein (CA)] and replication enzymes (protease, RT, and integrase) are produced during virus particle maturation via proteolytic cleavage of precursor Gag and Gag-Pol polyproteins, respectively (3). MA forms a spherical shell that interacts with the viral membrane (4, 5). Interior to the MA lattice lies the viral core. Roughly cone-shaped, the core is composed of the capsid outer shell, which itself is composed of CA. Core luminal components, which include two copies of the plus-stranded viral RNA genome, NC, RT, and integrase (6 – 8) (Fig. 1A), play critical roles during the early steps of HIV-1 replication.
Fig 1.
The HIV-1 core, CA, and capsomeres. (A) The drawing depicts the core with luminal components noted in text. IN, integrase. The approximate width of the wide end of the core is indicated. (B) Structure of the CA monomer (based on Protein Data Bank (PDB) accession code 4XFY) (9). The NTD, short interdomain linker, and CTD are colored forest green, magenta, and pale green, respectively. CYP-binding loop and FG pocket locations within the NTD are noted. N57 and N74 sidechains, which form part of the FG pocket, are drawn as sticks. (C) All-atom model of the capsid shell built from 186 hexamers and 12 pentamers (PDB accession code 3J3Y) (10) alongside resected CA hexamer [PDB code 4U0D (11)], pentamer [PDB code 3P05 (12)], and hexamer-2 [PDB code 6ECO (13)] structures. Hexamer and hexamer-2 colorings are the same as in panel B; pentamer-specific NTDs and CTDs are colored navy and light blue, respectively. Arg18 sidechains within the hexamer, which surround the R18 pore label, are shown as sticks. Coalesced NTD Glu75 and CTD Glu212 and Glu 213 residues are shown in space-fill at the tri-hexamer interface, where a single copy of each residue is labeled.
HIV-1 ingress begins with entry of the virus particle into a susceptible CD4-positive cell and terminates with formation of the provirus, which is an integrated DNA copy of viral RNA (diagrammed as steps 1–5 in Fig. 2). While unintegrated HIV-1 DNA can support some level of viral gene expression (14 – 18), recent research has clarified that heterochromatinization restricts the transcriptional capacity of retroviral DNA prior to integration (19 – 24). Provirus formation, accordingly, critically determines productive HIV-1 replication (14 – 16, 25, 26). The early events of HIV-1 replication are dedicated to cell entry (Fig. 2, step 1: Membrane fusion), reverse transcription and trafficking of the viral replication machinery from the cell periphery to the nuclear membrane (step 2: Cytoplasmic transport), nuclear import (step 3), intranuclear trafficking to preferred sites of integration (step 4), and, finally, integrase-mediated integration (step 5). Research conducted over the past several years has highlighted a central role for the capsid in mediating the steps of HIV-1 ingress that occur after cell entry (Fig. 2, steps 2–5). At the same time, the capsid helps to regulate the detection of HIV-1 nucleic acids by intracellular innate immune sensing machineries (27 – 33). Herein, we overview details of the capsid structure and common host factor binding elements within this structure. We then describe the myriad of cell factors that reportedly bind the capsid to aid its journey from the cell exterior to the nuclear interior. Along the way, we discuss potential experimental approaches to address lingering unknowns on the roles played by these host factors in HIV-1 ingress. We note several previous reviews that likewise discussed capsid–host interactions during the early steps of HIV-1 replication (34 – 42), some of which readers may wish to consult as background to the more-recently published works also presented here.
Fig 2.
Schematic of HIV-1 ingress. Following virus-cell membrane fusion (step 1), the viral core in association with motor complex adapter proteins such as BICD2 and FEZ1 travels along microtubules (step 2) toward the microtubule organizing complex (MTOC)/nuclear membrane. The interaction of the core with cell proteins such as CPSF6, NUP358, and NUP153 facilitates transport through the nuclear pore complex (NPC) (step 3). The CA-CPSF6 interaction further licenses core incursion into the nucleoplasm (step 4) toward speckle-associated domain (SPAD) regions of chromatin for integration (step 5). The leftward flow depicts steady-state condition in the absence of HIV-1 infection whereby the β-karyopherin transportin 3 (TNPO3) engages pre-mRNA splicing factors in the cytoplasm to affect their nuclear transport and subsequent downstream targeting of nuclear speckles (43, 44). LAD, lamina-associated domain; LEDGF, lens epithelium-derived growth factor; PIC, preintegration complex; Pol II, RNA polymerase II.
CAPSID STRUCTURE AND HOST FACTOR BINDING SITES
HIV-1 CA is an ~231-residue polypeptide that folds into two largely alpha-helical domains: the N-terminal domain (NTD) and C-terminal domain (CTD) (45, 46) (Fig. 1B). Independent CA molecules in turn interact intermolecularly to form distinct ring-like capsomeres, which are the building blocks of the capsid shell. The major capsomere is a hexamer of six CA molecules, while the minor capsomere is a CA pentamer (9, 12, 47, 48). The capsid shell is built from approximately 200 hexamers and exactly 12 pentamers (10, 49, 50) (Fig. 1C). The pentamers provide declinations to the otherwise regular hexameric honeycomb surface to induce curvature for shell closure. The distribution of seven pentamers at one end of the structure and five at the opposing end provides the cone shape typical of HIV-1 capsids (49).
Different types of in vitro assays are used by investigators to assess the binding of host factors to HIV-1 capsid, and it is worth noting that the nature of the binding assay can significantly influence the types of conclusions that can be drawn. Reactions performed with purified components, including proteins or peptides, can provide evidence for direct protein binding. Depending on reactants and techniques, affinity binding constants may be determined, and structure-based approaches such as NMR, X-ray crystallography, cryogenic electron microscopy (cryo-EM), or molecular dynamics (MD) may further yield atomistic models of bound complexes. Genetic-based approaches that then confirm the importance of the visualized protein-protein contacts in HIV-1 replication round out some of the most complete capsid–host interaction studies.
HIV-1 CA purified from recombinant sources such as Escherichia coli bacteria displays favorable biochemical properties, typically yielding protein concentrations as high as 20 mg/mL in isotonic salt-containing buffers. Early protein binding studies leveraged monomeric CA constructs including the isolated NTD (51 – 53). Determination of conditions that yielded the construction of stable CA hexamers (9, 47) subsequently led to their use in binding and structure-based studies, revealing novel contacts with the host that were missed from prior studies that utilized unassembled CA substrates (11, 54). Still, substrates that harbor the multi-hexamer honeycomb arrangement of the mature capsid lattice will capture interactions that exceed the binding capacities of individual CA molecules or capsomeres (Fig. 1C).
Recombinant CA-containing proteins under appropriate in vitro conditions template the formation of tube-like honeycomb arrays, a.k.a. capsid nanotubes, the surfaces of which mimic the surface of the mature capsid lattice (49, 55, 56). HIV-1 Gag is composed of MA-CA-spacer peptide 1 (SP1)-NC-SP2-p6 domains and recombinant CA-SP1-NC protein in the presence of templating nucleic acid formed nanotubes and capsid-like particles (CLPs) in isotonic salt conditions (49). These findings spearheaded the use of mature CA lattice-containing structures in host factor binding studies (53, 57 – 62). Because the comparatively large nanotubes/CLPs pellet by centrifugation at nominal g, host factor binding is oftentimes assessed via co-pelleting. Although CA on its own could also template nanotube formation (55), such structures required comparatively high salt concentration (1–2 M) to remain intact, limiting binding assays to nonphysiological salt conditions. Investigators have described two common workarounds of this bottleneck. One utilizes CA protein modified by cysteine replacement at positions 14 and 45 (A14C/E45C) (47), which, upon disulfide crosslinking, yields nanotubes that remain stable at isotonic salt conditions (63). The other advancement is inositol hexakisphosphate (IP6), which, since its discovery as a CA pocket factor, has significantly widened physiologically relevant approaches to capsid-host factor interaction and structure-based studies. At pH 8, unmodified recombinant CA in the presence of IP6 can form nanotubes that remain stable in isotonic salt buffers. Assembly conditions at pH 6 moreover yield a predominance of CLPs (48, 56). Leveraging such CLPs in single particle cryo-EM studies (48, 64) in the coming years will predictably revolutionize the field to yield a wealth of information on the structural details of HIV-1 capsid-host interactions. Complementary advancements in structural methodologies include cryo-electron tomography to visualize host factors bound to native HIV-1 cores following perforation of viral membranes (65).
On the host factor side, binding is oftentimes measured in the context of cell extracts, which fails to address whether host-capsid interactions are direct or perhaps require unknown intermediators. In these latter cases, the detected binding is indirect. This importantly contrasts results of direct protein binding that can be gleaned from work with purified components. Table 1 lists human proteins that have been reported to bind CA/capsid, most of which will be discussed in the main text, and whether these interactions have been shown to be direct. When known, the site/region of the capsid that mediates host binding is indicated. The table also lists the physical locations of the host factors within the cell. Following are descriptions of distinct aspects of HIV-1 CA and the capsid structure that serve as common host factor binding sites.
TABLE 1.
CA-interacting host factors for HIV-1 ingress
Ligand | UniProtKB | Intracellular localization a | Binding | Binding site | Reference(s) |
---|---|---|---|---|---|
BICD2 | Q8TD16 | Cytosol | Direct | – | (66, 67) |
CLASP2 | A0A804HJG7 | Cytosol | – | – | (68) |
CLIP1 | P30622 | Cytosol; microtubule ends | – | – | (69) |
CPSF6 | Q16630 | Nucleoplasm | Direct | FG pocket | (11, 54, 58, 70) |
CYPA | P62937 | Cytosol; nucleus b | Direct | CYP-binding loop | (51, 65, 71 – 73) |
CYPB | P23284 | Nucleus | Direct | CYP-binding loop | (71, 74) |
DAXX | Q9UER7 | Nucleus; nuclear bodies | – | – | (75) |
DCTN1 | Q14203 | Actin filaments; cytosol | – | – | (76) |
DIAPH1 | O60610 | Plasma membrane | – | – | (77) |
DIAPH2 | O60879 | Nucleoli; ER | – | – | (77) |
FEZ1 | Q99689 | Cytosol | Direct | R18 pore | (78, 79) |
MAP1A | P78559 | Cytosol | – | – | (80) |
MAP1S | Q66K74 | Cytosol | – | – | (80) |
MAPK1 | P28482 | Cytosol; NS c | – | – | (81) |
MARK2 | Q7KZI7 | Nucleoplasm; PM | – | – | (82) |
MELK | Q14680 | Cytosol; nucleus d | – | – | (83) |
MX2 | P20592 | Nuclear membrane e | Direct | Tri-hexamer interface | (61, 62, 84 – 87) |
NONO | Q15233 | Nucleoplasm | Direct | HIV-2 CA residues 49, 101, 102 | (33) |
NUP153 | P49790 | Nuclear membrane | Direct | FG pocket; tri-hexamer interface | (11, 53, 54, 60, 88, 89) |
NUP214 | P35658 | Nucleus | – | – | (59, 88, 90) |
NUP358 | H2QII6 | Nuclear membrane; vesicles | Direct | CYP-binding loop | (91, 92) |
NUP62 | P37198 | Nuclear membrane; vesicles | Direct | – | (88, 93, 94) |
NUP88 | Q99567 | Nucleoplasm | – | – | (93) |
NUP98 | P52948 | Nucleoplasm | – | – | (60) |
PDZD8 | Q8NEN9 | Nucleoli; PM | – | – | (95) |
PIN1 | Q13526 | Nucleoplasm; cytosol | – | – | (96) |
PQBP1 | O60828 | NS | Direct | – | (31) |
SEC24C | P53992 | Vesicles | Direct | FG pocket | (90) |
SUN1 | O94901 | Nuclear membrane | – | – | (97, 98) |
SUN2 | Q9UH99 | Nuclear membrane | – | – | (97, 98) |
TNPO1 | Q92973 | Nucleoplasm; cytosol | Direct | CYP-binding loop | (99) |
TNPO3 | Q9Y5L0 | Vesicles | – | – | (100) |
TRIM11 | Q96F44 | Nucleus; cytosol | Direct | – | (101) |
TRIM34 | Q9BYJ4 | Nucleoli; centrosome | – | – | (102) |
TRIM5α | Q9C035 | Cytosol | Direct | Mature lattice | (103 – 105) |
The CYP-binding loop
Cyclophilin (CYP) A and B, encoded by human PPIA and PPIB genes, respectively, were initially shown using the yeast two-hybrid assay to interact with HIV-1 Gag protein (71). CYPA was subsequently shown to directly bind an exposed loop within the CA NTD (the CYP-binding loop) (45, 51) (Fig. 1B). One consequence of the Gag-CYPA interaction is the incorporation of the host factor into virus particles (111, 112). However, subsequent research revealed that the interaction with the CYPA protein present in nascently infected target cells, as compared to the CYPA in virus particles, determines the host factor’s role in HIV-1 infection (113, 114).
The CA-CYPA interaction can impact many steps throughout the early phase of the virus lifecycle, including cytoplasmic trafficking (107), nuclear import (115), and integration site targeting (52). Recent work has shown that CYPA can regulate the interaction of CA with other host factors, including cleavage and polyadenylation specificity factor 6 (CPSF6) (107), Sad1 and UNC84 domain containing (SUN) 1 (116), SUN2 (117), tripartite motif 5α (TRIM5α) (118, 119), and myxovirus resistance 2 (MX2) (120). Parallel studies have identified within the mature capsid lattice two CYPA binding sites in addition to the canonical CYP-binding loop (72, 73). Plausibly, the ability to regulate the interaction of several host factors with the capsid underlies the multifaceted role of CYPA in HIV-1 ingress.
Although much less studied than CYPA, the CA-CYPB interaction has also been shown to regulate HIV-1 nuclear import (74). All told, there are more than 20 human proteins with predicted cyclophilin-like domains, 7 of which can be depleted from cell extracts using glutathione S-transferase (GST)-tagged HIV-1 Gag in a pulldown assay format (121). The CTD of the large nucleoporin (NUP) NUP358 (a.k.a. RAN binding protein 2 or RANBP2), which is a component of the nuclear pore complex (NPC), contains a cyclophilin-homology (CypH) domain that engages the CYP-binding loop similarly as CYPA (52, 91, 92). Additional work is required to determine if cyclophilin domain-containing human proteins beyond CYPA, CYPB, and NUP358 play a role in HIV-1 ingress and, if so, where along the infection pathway they may exert their affects.
The Phe-Gly binding pocket
CPSF6 (11, 54, 70), NUP153 (11, 53, 54, 88), and SEC24 homolog C (SEC24C) (90) use specific Phe-Gly (FG) peptides to engage the FG-binding pocket (Fig. 3A). Although formulated primarily via the CA NTD (Fig. 1B), CTD elements from an adjacent CA molecule within the capsomere ring contribute important elements to the FG pocket (11, 54, 88, 90, 122). Recent research indicates that CA hexamers as compared to pentamers harbor functional FG-binding pockets (48, 123).
Fig 3.
Host factor binding sequences and regions. (A) Domain arrangements of FG host factors NUP153, SEC24C, and CPSF6. Short vertical lines mark positions of FG peptides, with green arrows highlighting specific FGs for capsid binding. Yellow shade demarcates PrLDs as predicted from the PLAAC server (124). The red ticks at the NUP153 C-terminus demarcate the terminal RRRKCOOH sequence (triple-Arg binding motif underlined). The CPSF6 PrLD lies amid a larger Pro-rich domain (PRD), which is denoted as orange rectangle. ZnF, zinc-finger domain; RRM, RNA recognition motif; RSLD, arginine-serine like domain [a.k.a. Arg-mixed charge domain or R-MCD (125)]. (B) Sequence alignment of similar stretches of FEZ1 (top) and PQBP1 (bottom) proteins. Numbers refer to amino acid positions within the respective proteins. Asterisks demarcate FEZ1 residues shown by MD to bind CA Arg18 at >87.8% occupancy, with E182 and E183 showing near complete occupancy (>99.5%). Black and gray backgrounds indicate identical residues and physiochemically conserved side chains, respectively.
While CPSF6 contains but a single FG peptide, NUP153 and SEC24C contain 29 and 8, respectively. Most of these sequences are located within the C-terminal FG domain of NUP153 and the NTD of SEC24C (Fig. 3A). Remarkably, despite the comparatively high density of FG peptides within the NUP153 FG domain, capsid binding maps to a single FG sequence (53, 89). Recent work has shown that the key capsid-binding FG sequences within CPSF6, NUP153, and SEC24C reside within prion-like domains (PrLDs) (122) (Fig. 3A, yellow), which are low-complexity regions (LCRs) devoid of charged amino acid residues (124). PrLD sequences abutting the FG peptides moreover play critical roles in high-affinity capsid binding. Thus, while ~15 residue synthetic FG peptides bound recombinant CA hexamers comparatively weakly (K d ~70 µM to 1 mM), the corresponding GST-tagged PrLD proteins bound purified HIV-1 cores with K d ’s in the 0.3–0.9 µM range. The structural basis of the CPSF6 PrLD-capsid interaction was visualized at the comparatively low resolution of ~7.9 Å using CA nanotubes and single-particle cryo-EM, which revealed that the FG-anchored PrLDs interlinked intermolecularly to form chains of CPSF6 molecules between adjoined rows of CA hexamers (122). These data suggest that a specific type of FG/PrLD-mediated higher-order interaction underlies the structural basis for avid FG host factor interactions with the HIV-1 capsid. Further details of the CPSF6-capsid interaction will be enumerated by studying larger CPSF6 substrates that incorporate the host factor’s N-terminal RNA recognition motif (RRM) and/or C-terminal Arg-Ser-like domain (RSLD) (Fig. 3A) and/or by improving the resolution of the cryo-EM maps. Parallel structure-based work with the PrLD segments of NUP153 and SEC24C will reveal if these domains form similar higher-order linked chains as observed for CPSF6.
SEC24C was identified via a mass spectrometry (MS)-based screen for capsid binding host factors using recombinant CA nanotubes as bait (90). Interestingly, this screen identified several additional host factors with FG sequences amid predicted PrLDs. Some of these proteins, such as NUP62, NUP98, and NUP214, were previously shown to co-pellet with CA-SP1-NC (60, 88) or CA nanotubes (93) in vitro. Additional work is required to determine if PrLD-embedded FGs are responsible for these host-capsid interactions, as well as for the interactions detected for other FG-containing host factors uncovered in the CA nanotube binding screen (90). Plausibly, the FG pocket mediates the binding of a comparatively large array of host factors for HIV-1 ingress.
The R18 pore
A locally enriched electropositive pore composed of Arg18 residues at the center of hexameric and pentameric capsomeres provides a binding platform for electronegative cellular components including relatively small metabolites and proteins (Fig. 1C). The binding of dNTPs to the R18 pore can facilitate their transport into the core for reverse transcription (126, 127). IP6 is incorporated into virions via engaging CA residues Lys158 and Lys227 in the context of HIV-1 Gag. Following maturation, IP6 is coordinated by the guanidino sidechain groups of the R18 pore (56, 128 – 130). Recent research has highlighted that IP6 mediates capsid assembly via kinetically trapping pentamers into growing hexameric lattices (131). As previously mentioned, its addition to CA assembly reactions at pH 6 yields a predominance of CLPs. Adding IP6 to permeabilized virions (132) or isolated cores (133) moreover dramatically stimulated the synthesis of double-stranded DNA in so-called endogenous reverse transcription assays, yielding DNA products that were competent for integration (132). These assays hold great promise to reconstitute various aspects of HIV-1 ingress under acutely controlled in vitro conditions.
The kinesin adapter protein fasciculation and elongation factor zeta 1 (FEZ1) binds capsid to effect HIV-1 ingress (78) (discussed in detail in the capsid-host interactions for cytoplasmic transport section). MD simulations revealed that FEZ1 leverages a stretch of electronegative residues, namely Asp180, Glu182, Glu183, and Glu184, to engage the capsid via the R18 pore (79). Polyglutamine binding protein 1 (PQBP1) engages HIV-1 capsid to effect innate immune sensing via cyclic GMP-AMP synthetase (cGAS) (31, 134). The N-terminal 46 residues of PQBP1, which, like FEZ1, is enriched in electronegative residues (Fig. 3B), interacted with capsids in cells more robustly than did the C-terminal fragment composed of residues 47–265 (31). Moreover, binding was effectively negated by the R18G amino acid substitution in CA. Additional structure-based work is required to determine if PQBP1 electronegative residues analogous to those in FEZ1 directly engage the R18 pore.
Restriction factors and the tri-hexamer interface
The main treatise of this paper is CA–host interactions that promote HIV-1 infection. Cells additionally harbor so-called restriction factors, which can thwart retroviral replication at distinct steps of their replication cycles [see references (135) and (136) for recent reviews]. Because some restriction factors, namely TRIM5α from rhesus macaques (137), DAXX (138), TRIM11 (101), TRIM34 (102), and MX2 (139, 140), can inhibit HIV-1 ingress via engaging the incoming capsid (57, 61, 62, 75, 84, 85), we, for completeness, include brief descriptions of these CA-binding protein functionalities.
TRIM5α forms a higher-order hexagonal lattice encasing the HIV-1 capsid (103, 104), the consequences of which accelerate uncoating and impede reverse transcription (57, 137). While TRIM11 also restricts reverse transcription via accelerating uncoating (101), DAXX reportedly impedes HIV-1 DNA synthesis via stabilizing incoming cores (75). MX2 restricts HIV-1 ingress a bit further down the infection pathway. While reverse transcription proceeds normally, MX2 restricts the accumulation of nuclear forms of HIV-1 DNA (unintegrated and integrated), indicating that MX2 primarily inhibits nuclear import or destabilizes post-import viral DNA (139 – 141).
MX2 is a multidomain protein composed of an N-terminal unstructured region, followed by GTPase and stalk domains with interspersed bundle signaling elements [reviewed in reference (142)]. While protein oligomerization (61, 62, 85) and the GTPase domain (86) contributed to capsid binding, binding is principally determined via an 11RRR13 triplet of arginine residues that reside near the protein’s N-terminus (87). MD simulations have shown that these Arg residues can interact with the electronegative side chain-enriched tri-hexamer interface that is formed via the juxtaposition of three neighboring CTDs from three distinct CA hexamers (13, 143) (Fig. 1C). Recent work has implicated a triplet of arginine residues near the C-terminus of Nup153—1472RRR1474 (Fig. 3A)—in capsid binding and HIV-1 nuclear import (89). MD simulations moreover indicated that this RRR motif—similar to the one in MX2—engages the tri-hexamer interface. Thus, while the main FG binding determinant of NUP153 engages individual CA hexamers, two additional protein elements—the PrLD and C-terminal RRR motif—potentially engage in higher-order interactions to impart high-affinity NUP153 binding to capsid. Additional research is required to determine the extents that the PrLD versus RRR motif contribute to avid NUP153-capsid interactions.
CAPSID-HOST FACTOR INTERACTIONS DURING HIV-1 INGRESS
Following are the descriptions of the roles purportedly played by CA-interacting proteins in capsid cytoplasmic trafficking, nuclear transport, and intranuclear trafficking to sites of viral DNA integration. A wide variety of approaches are used to assess the roles of host cell factors in HIV-1 replication. With the advent of RNA interference (RNAi) in 2001 (144), researchers could effectively deplete specific host factors from mammalian cells to assess their influences on virus infection and replication. The subsequent application of CRISPR-Cas9 has significantly expanded ways in which researchers can deplete specific mammalian cellular factors (145). The advent of complementary knockdown technologies moreover helps to alleviate concerns that may arise from technique-specific off-target effects. If the targeted factor is nonessential for cell viability, CRISPR-Cas9 can further be used to generate knockout cell lines. HIV-1 mutant viruses with changes in CA residues that are known to effect capsid-host interactions are also commonly used in the field. Because any given CA change might affect interactions with more than one cell factor (see preceding section), comprehensive studies often combine the study of CA mutant viruses with RNAi/CRISPR-Cas9 depletion strategies.
HIV-1 capsid uncoating
Because measures of capsid uncoating are commonly employed in HIV-1 ingress studies, we would be remiss without a short overview of this subject. We by no means intend this to be comprehensive—readers are directed elsewhere for reviews dedicated to HIV-1 uncoating (146 – 148).
Operationally, uncoating refers to loss of CA or capsomeres (or perhaps larger chunks) from the capsid shell during HIV-1 ingress. Both biochemical and microscopy-based approaches have been used to assess uncoating. Two intermediates of retroviral early event replication, namely, reverse transcription complexes (RTCs) (149, 150) and preintegration complexes (PICs) (151 – 153), are defined as high-molecular weight entities in cell extracts that support the processes of reverse transcription and integration, respectively. Initial analyses of partially purified HIV-1 RTCs and PICs by western immunoblotting revealed little, if any, CA content (150, 154, 155). Evidence for CA among active PIC fractions was subsequently revealed using more sensitive enzyme-linked immunosorbent assays and immunoprecipitation techniques (156). The fate-of-the-capsid (FOC) assay, which is a state-of-the-art biochemical uncoating assay, leverages the pelletability of CA soon after cell lysis. Intact capsids or other high molecular weight CA structures partition to the pelleted fraction while uncoated CAs (individual molecules or perhaps capsomeres) remain in supernatant fractions (57, 157). FOC assays are routinely used in studies that test the effects of specific host factors in uncoating. Advanced biochemical approaches that leverage fluorescent microscopy to monitor CA content of individual, permeabilized virions attached to glass slides have more recently been used to assess uncoating (158, 159).
The advent of advanced cell biology techniques (160) opened the door to in situ observation of replication intermediates in intact cells to assess HIV-1 uncoating. Numerous approaches, including accessibility of labeled RNA after virus-cell fusion (161) or loss of fluorescent marker proteins such as encapsulated, fluid phase green fluorescent protein (GFP) (162, 163), have been utilized. Systems that track loss or fluorescent fusion proteins with CA (164 – 166) or CYPA (167, 168) have also been used, though fluorescent tagging of important ingress factors such as CA or CYPA may subtly alter protein function. Recent attempts to mitigate potential off-targeting affects have leveraged the comparatively noninvasive labeling technique known as genetic codon expansion, where unnatural amino acid residues can be labeled in situ via click chemistry (169). Another confounding issue for ingress studies is that less than half of HIV-1 particles are infectious (170), necessarily complicating microscopy-based approaches. One ingenious workaround to this caveat is end-point dilution, whereby integration-dependent viral reporter genes are used to earmark cells productively infected via single viral particles (162, 168).
Different uncoating scenarios can be envisioned during HIV-1 ingress [comprehensively reviewed in reference (171)]. One posits that complete uncoating occurs soon after cell entry. While not uncommon in the early literature, the advent of microscopy-based approaches has since de-popularized this theory. Another posits progressive loss of CA during ingress, with perhaps nuclear transport requiring near-complete uncoating. This stance was in part based on the long-held view that the width of the wide end of the HIV-1 core, ~60 nm (Fig. 1A) (50), exceeded the ~40–45 nm width of the NPC inner channel (172). However, recent work based on images from intact cells has indicated greater plasticity in NPC organization than once thought. In particular, the average inner diameter of 64 nm in T cells was sufficiently large to accommodate HIV-1 capsids for nuclear transport (173). These observations are consistent with other microscopy-based findings that HIV-1 capsids can remain largely intact during nuclear transport (163, 165, 169), with perhaps some remodeling required to pass through the nucleocytoplasmic sieve (166, 174, 175). Recent findings that nuclear factors such as CPSF6 and NUP153 (89, 122) display high affinity binding to mature capsid lattice substrates is consistent with the notion that some aspect of the mature lattice must survive nuclear transport and that HIV-1 uncoating primarily occurs post nuclear import (148, 163, 165, 169, 176, 177).
A key driving factor of HIV-1 uncoating is the physical process of reverse transcription (178 – 183). Indeed, imaging reconstituted endogenous reverse transcription reactions that also support integration has revealed fenestrations where apparent nascent DNA strands extrude from broken or cracked capsid shells (132). Such observations raise the intriguing possibility that integration may proceed before the capsid shell is fully disassembled.
Capsid-host interactions for cytoplasmic transport
The makeup of the intracellular environment, which is heavily populated by macromolecular complexes, precludes directional inward movement of large entities such as viruses or subviral complexes by passive diffusion [reviewed in reference (184)]. Viruses accordingly have evolved to hijack intracellular transport systems to navigate the cellular milieux. The cell harbors a cytoskeleton composed of actin microfilaments (~7 nm in diameter), intermediate filaments (~10 nm), and microtubules (~25 nm) [reviewed in reference (185)]. Microtubules are dynamic structures composed of α-tubulin and β-tubulin that enable the assisted trafficking of cargoes inward toward the nucleus (retrograde movement) or outward toward the cellular periphery (anterograde movement). The orientation of tubulin dimers within the polymers provides microtubules with intrinsic polarity. Microtubule plus-ends localize toward the cell periphery, while the minus-ends congregate with the microtubule organizing complex (MTOC), a comparatively large structure that, during interphase, often associates with the nuclear membrane (Fig. 2) [reviewed in reference (186)]. Different types of molecular motor proteins, namely dynein and kinesins, associate with microtubules to facilitate retrograde versus anterograde transport, respectively [see reference (187) for review].
Microtubules can become locally stabilized, which is associated with specific α-tubulin post-translational modifications including acetylation and detyrosination (188). HIV-1 infection induced the formation of stable microtubules, which required the function of microtubule plus-end tracking proteins (+TIPs) including microtubule-associated protein (MAP) RP/end-binding (EB) family member 1 (MAPRE1; a.k.a. EB1) (189), diaphanous-related formin 1 (DIAPH1), and DIAPH2 (77). While CA-SP1-NC nanotubes depleted DIAPH1 and DIAPH2 from cell extracts in vitro, MAPRE1 has not revealed capsid binding activity (69, 77). Interestingly, the sequence of the MAPRE1 C-terminal region mimics the sequence of the N-terminal portion of the HIV-1 CA CTD, which coincides with the phylogenetically conserved major homology region (69, 190). Thus, CA has seemingly evolved to mimic MAPRE1 to congregate +TIPs for effective microtubule engagement. Indeed, other +TIPs, namely cytoplasmic linker protein (CLIP)-associated protein 2 (CLASP2) (68) and CLIP1 (a.k.a. CLIP170) (69), have been shown to facilitate HIV-1 cytoplasmic transport and bind HIV-1 CA-SP1-NC nanotubes in cell extracts. MAPs MAP1A and MAP1S also facilitate microtubule stabilization and can interact with HIV-1 cores to affect their retrograde transport (80). Stabilized microtubules provide tracks for comparatively long-range motility, which is exploited by HIV-1 for the inward movement of its core.
HIV-1 capsids bind specific adapter proteins to engage ATP-metabolizing motors for microtubule-dependent transport. Eukaryotic cells harbor numerous kinesins but only one type of cytoplasmic dynein, which, during interphase, plays critical roles to traffic membrane-bound organelles, RNAs, protein complexes, and viruses [reviewed in reference (191)]. Dynein is a 13-component protein complex composed of heavy chains DYNC1H1 and DYNC2H1, intermediate chains DYNC1I1 and DYNC1I2, and various light intermediate and light chains. Dynein function requires the co-factor dynactin, which itself is a multi-component complex. Adapter proteins, which typically harbor coiled-coil domains, tether cargoes to the dynein-dynactin supracomplex to effect directional movement (191). The adapter protein bicaudal D homolog 2 (BICD2) enhanced the affinity of the dynein-dynactin interaction and dramatically activated the motility of the macromolecular complexes to move long distances in vitro (192, 193). BICD2 effectively bound CA/CA-SP1-NC nanotubes in vitro and was required for optimal HIV-1 infection and cytoplasmic trafficking (66, 67). By using purified recombinant BICD2 proteins, the interaction with capsid was moreover shown to result from direct binding (67). Several dynein/dynactin proteins in cell extracts also co-pelleted with CA-SP1-NC/CA nanotubes in vitro. Because the binding of these proteins to capsid lattices depended on the cellular concentration of BICD2, dynein/dynactin protein binding was almost certainly indirect and mediated via BICD2 (67) (Table 1). A separate study determined that the dynactin component DCTN1 can act as a +TIP to compete with CLIP1 for binding to HIV-1 cores and accordingly negatively regulate microtubule engagement and viral infection (76).
Kinesins comprise a comparatively large superfamily of ~45 related genes in mice that can be subclassified into 15 families [reviewed in reference (194)]. Based on the relative location of kinesin motor domains, family members can be broadly typed into N-kinesins (N-terminally located motors), M-kinesins (midregion located motors), and C-kinesins (C-terminal motors). While M-kinesins typically support retrograde transport of cargoes, N-kinesins typically support anterograde transport (194). Indeed, early studies implicated N-kinesins KIF4 (195, 196) and KIF3A (197) in Gag transport for retroviral assembly and release from cells. Perhaps counterintuitively, N-kinesins have also been implicated in retrograde transport of the HIV-1 core.
Depletion of cellular N-kinesin KIF5B was initially shown to delay the process of capsid uncoating in HIV-1-infected cells (198). Akin to dynein-dynactin, adapter proteins tether kinesin motor proteins to cargoes, and the kinesin adapter protein FEZ1 was subsequently shown to co-pellet with CA-SP1-NC nanotubes and facilitate the inward movement of HIV-1 cores (78). Purified FEZ1 proteins specifically interacted with CA hexamers and, as discussed above, MD simulations have yielded an atomistic model of electronegative FEZ1 residues that engage the R18 pore (79). Phosphorylation of FEZ1 by microtubule affinity-regulating kinase 2 (MARK2) in association with HIV-1 cores enhanced FEZ1’s interaction with KIF5B and facilitated retrograde transport and HIV-1 infection (82). KIF5B moreover induced the relocalization of NUP358 from the nuclear membrane to the cytosol in a manner that was dependent on the capsid-CPSF6 interaction (199). Concordantly, capsid-CPSF6 complexes have been observed to traffic along microtubules (107). Plausibly, the connection of KIF5B with downstream factors such as NUP358 and CPSF6, which are predominantly associated HIV-1 nuclear import and intranuclear trafficking, respectively (199 – 203), instills overall retrograde movement for a kinesin that would usually move cargoes anterogradely. It also seems possible that KIF5B/FEZ1 could support the anterograde transport of HIV-1 cores from the MTOC to proximal NPCs for nuclear transport (Fig. 2) (199).
SEC24C is a member of the coat protein complex II or COPII complex that typically plays a role to transport cargoes from the endoplasmic reticulum to the Golgi apparatus (204), though a separate COPII component, SEC13, forms part of the NPC (205, 206). As described above, SEC24C was uncovered in an MS-based screen for host factors that interact with CA nanotubes (90). Knockout cells revealed a critical role for SEC24C in the production of HIV-1 under multi-cycle replication conditions. When analyzed under single round infection conditions, HIV-1 displayed similar 2- to 3-fold defects in core stability, reverse transcription, and nuclear import, indicting an important role for this FG-pocket factor to maintain optimal core stability during HIV-1 ingress (90).
Phosphorylation can also reportedly regulate capsid stability during the early stage of HIV-1 infection. Maternal embryonic leucine zipper kinase (MELK) phosphorylates CA residue Ser149 for optimal reverse transcription and core uncoating (83). Peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1) is a phosphopeptide-binding enzyme that isomerizes phosphorylated (S/T)-P peptide bonds (207). Virion-associated mitogen-activated protein kinase 1 (a.k.a. ERK2) phosphorylation of CA residue Ser16 was critical for the association of PIN1 with capsid and optimal core uncoating and HIV-1 infection (81, 96).
PDZ domain-containing protein 8 (PDZD8) was identified through a yeast 2-hybrid screen to interact with HIV-1 Gag and to play an important role in virus infection (208). PDZD8 in cell extracts was subsequently shown to co-pellet with CA-SP1-NC nanotubes, and RNAi-mediated knockdown reduced HIV-1 infection via accelerating the rate of capsid uncoating, which negatively impacted reverse transcription (95). Perhaps unexpectedly, HEK293T and HeLa cell clones knocked out for PDZD8 expression supported the wild-type levels of virus infection, indicating that PDZP8 is not an essential HIV-1 host cofactor (209).
The NPC and HIV-1 nuclear import
Nucleocytoplasmic transport is regulated by the NPC, a massive assembly of NUPs that precludes passive diffusion of comparatively large macromolecules (210). Eukaryotic cells have accordingly evolved specific chaperon-like nuclear transport receptor (NTR) proteins, also called β-karyopherins, to assist in the nucleocytoplasmic transport of comparatively large cellular cargos [reviewed in reference (211)]. Occasionally, cellular cargoes can access the NPC through direct binding of one of its constitutive components (212).
The human NPC is constructed from ~33 NUPs arranged in 8-fold rotational symmetry [reviewed in reference (206)]. Various NUP subcomplexes, namely cytoplasmic NUPs, the Y-complex, inner ring NUPs, pore membrane proteins, and nuclear basket NUPs, come together to form the NPC. Of the NUPs that have been shown to interact with CA (Table 1), NUP358, NUP214, and NUP88 are cytoplasmic filament NUPs, NUP62 and NUP98 are inner ring NUPs, and NUP153 is a nuclear basket NUP. Nine of the 33 human NUPs contain FG-repeat domains, including NUP214, NUP62, NUP98, and NUP153. The molecular sieving property of the NPC is determined by conglomeration of NUP FG-repeat domains within the inner NPC channel (206). It is important to note that capsid binding activity does not necessarily translate to a role for the NUP protein in HIV-1 nuclear import. For example, NUP214 function has been mapped to the comparatively late step of HIV-1 mRNA nuclear export (59).
The HIV-1 capsid has evolved specific interactions with NPC components to affect its nuclear transport. The interaction with NUP358 likely docks HIV-1 cores at the NPC (52, 59, 199). Although capsid binding predominantly maps to the RBD4-CypH didomain composed of the C-terminal CypH and adjacent Ran-binding domain IV (52, 59, 94), CA-SP1-NC in cell extracts co-pelleted a C-terminal truncation mutant lacking these domains about 40% as efficiently as full-length NUP358 (59). Plausibly, this residual binding activity accounts for CypH-independent NUP358 functionality under certain conditions of HIV-1 infection (213). Although not classified as possessing an FG-repeat domain, NUP358 does harbor 22 FG dipeptides, including 19 upstream of the C-terminal didomain. None of these, however, is predicted to lie within a PrLD. Additional work is required to map NUP358 determinants upstream of the RBD4-CypH didomain that contributes to capsid binding.
Binding reactions conducted with purified proteins have shown that NUP358 and NUP153 interact directly with CA (52, 53). HIV-1 nuclear import is moreover impeded in NUP358 or NUP153-depleted cells (52, 59, 200, 214). The β-karyopherin transportin 1 (TNPO1 or TRN-1) has also been shown to directly bind CA (99). While this study revealed significant HIV-1 uncoating and nuclear transport defects in TNPO1-depleted cells, other studies reported no obvious HIV-1 infection defect in cells depleted for TNPO1 (93, 215). Additional work is accordingly warranted to investigate the contribution of the CA-TNPO1 interaction to HIV-1 nuclear import.
The β-karyopherin TNPO3 (a.k.a. TRN-SR2) has also been shown to play a significant role in HIV-1 nuclear import (216). Although first thought to be mediated via an interaction with integrase, subsequent work showed that the requirement for TNPO3 during HIV-1 infection mapped to CA and not integrase (217). Additional work has indicated that the connection between TNPO3 and CA is predominantly indirect and mediated via CPSF6 (218, 219). CPSF6, which is a member of the cleavage factor I mammalian (CFIm) complex (220), was first implicated in HIV-1 biology via the ability of a C-terminal truncation variant to potently restrict HIV-1 infection and nuclear import (58). Subsequent work revealed that TNPO3 is the β-karyopherin responsible for CPSF6 nuclear import (221, 222). In this way, TNPO3 depletion partially relocalized the normally nuclear sequestered CPSF6 to the cytosol, the consequences of which restricted HIV-1′s access to the nucleus (218, 219). Although CA-SP1-NC could deplete TNPO3 from cell extracts (100), this binding may very well be mediated via CPSF6. Additional work with CPSF6 knockout cells (223) and purified TNPO3 protein (221) could help to clarify if TNPO3 binds CA in a CPSF6-independent manner. Follow-up work has confirmed a marginal role for the TNPO3-integrase interaction in HIV-1 nuclear import (224, 225).
The role of CPSF6 in HIV-1 infection is highly context dependent, in general requiring spreading replication assays with primary cells to see substantial effects. For example, HEK293T knockout cells, if anything, supported increased levels of HIV-1 single-round infection compared with control cells (223), similar to responses observed using other transformed cell types depleted for CPSF6 expression via RNAi (58, 226). While RNAi-mediated knockdown in primary macrophages yielded an approximate 2.5-fold infection defect under single round conditions (203), this approach in a separate study counteracted the ability for the cells to support spreading HIV-1 replication (29). CRISPR-Cas9-mediated depletion of CPSF6 from primary, resting CD4+ T cells moreover reduced HIV-1 infection ~7- to 10-fold under single round conditions (227). Approximate 2- to 3-fold reductions in HIV-1 nuclear import have been reported under certain CPSF6 knockdown conditions (201, 203).
An ingenious approach to the study of HIV-1 nuclear import leveraged a drug inducible forced dimer construct of NUP62, called Nup62DG (176). By staggering the time of drug addition, these investigators were able to assess the time required post-infection to achieve 50% nuclear import, which ranged from about 3–4 h in CD4+ T cells and macrophages to about 7 h in HeLa cells. The resistance of the P90A CA mutant virus, which is defective for CYPA binding (111), to the effects of Nup62DG (176) suggests a potential cyclophilin component to NUP62 dependency during HIV-1 infection. Additional work with factor-specific knockdowns under baseline infection and drug-induced Nup62DG restricted conditions could potentially flush out such a dependency.
Recent in vitro work with DNA-origami mimics of the human NPC (called NuPODs for NUPs organized by DNA) suggests a potential model for HIV-1 nuclear import (94). The NuPOD scaffold afforded specific placement of NUP domains, or in the case of NUP62, the full-length protein, in regions representative of the NPC cytoplasmic face (NUP358 RBD4-CypH didomain), nuclear face (NUP153 FG domain), or in between (NUP62). The NUP153 FG domain revealed strong preference for binding to comparatively curved regions of the capsid lattice, including the narrow “tip end” of HIV-1 cores that feature five capsomere pentamers (Fig. 1C). Based on co-pelleting of purified proteins with CA nanotubes, the NUP153 FG-repeat domain bound capsids at higher affinity than did either the NUP358 RBD4-CypH didomain or full-length NUP62 protein. Accordingly, after NUP358-mediated docking at the NPC, an apparent binding affinity gradient from the cytoplasmic side of the NPC toward the nucleoplasm, combined with the affinity of the NUP153 FG domain for the highly curved tip, would naturally orient the core for tip-first insertion and passage through the NPC. While the tip-first model is consistent with some tomographic images of HIV-1-infected cells (173), there are several caveats with the NuPOD system that deserve further testing. First, while the FG domain seemingly fully accounts for NUP153’s capsid binding activity (53), elements upstream of the RBD4-CypH didomain, as mentioned, contribute to the NUP358-capsid interaction (59). Larger versions of NUP358 need to be tested to confirm if indeed NUP153 binds HIV-1 capsids with higher affinity than does (full-length) NUP358. Second, although CA-containing nanotubes readily interacted with NUP62 in cell extracts (88, 93) and with purified NUP62 protein (94), they failed to engage NUP62-engrated NuPODs. Moreover, engrafted NUP62 presented a barrier that HIV-1 cores could not effectively breach (94). Numerous FG NUPs, including NUP62, have the ability to form hydrogels (228), and it seems possible that multiple NuPOD-engrafted copies surpassed the local NUP62 concentration required for hydrogel formation. How then could the HIV-1 core breach cellular NPCs? In cells, NUP62 forms the channel NUP heterotrimer (CNT) complex with NUP54 and NUP58 partner proteins (229), so reconstituted CNTs should be tested in place of NUP62 to see if the physiological trimeric complex might present less of a barrier than did engrafted NUP62. Tests with relevant β-karyopherins, including TNPO1 and/or TNPO3 (± CPSF6), could also be performed, as the physiological roles of such NTRs is to ferry cargoes through the NPC. Finally, HIV-1 infection can induce the relocalization of nuclear-membrane bound NUP62 to the cytoplasm to associate with HIV-1 cores (176, 230), indicating that viral infection may help to reduce the barrier-forming properties inherent to the inner NPC channel. Additional experiments should be performed to test whether the other CNT proteins are also similarly relocalized in response to HIV-1 infection.
Nuclear trafficking and HIV-1 integration
An over-expression screen for nuclear envelop-associated proteins identified SUN1 and, to a lesser extent, SUN2, as potent inhibitors of HIV-1 infection. Sensitivity to SUN1 mapped to CA, and SUN1 and SUN2 in cell extracts co-pelleted with CA-SP1-NC nanotubes in vitro. While SUN2 knockout yielded marginal ~2-fold defects in HIV-1 infection, there was no apparent infection defect using SUN1 knockout cells (97). While restriction of infection by over-expression and CA-binding activities are reproducible across studies (98, 231), the physiological roles of SUN1 and SUN2 in HIV-1 infection await clarification. One RNAi-based study indicated that integration was significantly impaired in SUN1 knockdown cells, though this defect was not associated with a parallel defect in HIV-1 infection (116).
CA-binding proteins can significantly influence sites of HIV-1 integration. HIV-1 integration into human DNA occurs nonrandomly, with preferences observed at multiple levels that span from local nucleotide sequence to global chromatin architecture [see references (232) and (233) for recent reviews]. HIV-1 integration displays marked preferences for annotations associated with active chromatin including genes, transcriptional activity, gene-dense regions (234), activating epigenetic marks (235), speckle-associated domains (SPADs) (236), and topologically associating domains (TADs) (237). As a consequence, integration significantly negatively correlates with heterochromatic markers including repressive epigenetic marks (235) and lamina-associated domains (202, 238), which locate outward toward the nuclear periphery in association with lamin proteins (239) (Fig. 2 and 4).
Fig 4.
Roles for the CA-CPSF6 interaction in HIV-1 intranuclear trafficking and integration targeting. (A) In the absence of its interaction with CA, CPSF6 localization remains largely pan-nuclear, HIV-1 trafficking arrests at the nuclear pore complex (NPC), and integration occurs at proximal chromosomal locations including lamina-associated domains (LADs). (B) The CA-CPSF6 interaction is critical for nuclear evasion of the HIV-1 core, CPSF6 puncta formation, and SPAD-integration targeting. Recent work has raised the intriguing possibility that CPSF6 LLPS activity underlies HIV-induced puncta formation (240), which will require additional experiments to formally prove or disprove. NS, nuclear speckles; SPAD, speckle-associated domain.
Gene knockout and RNAi studies have highlighted two virus-host interactions that in large part account for HIV-1 integration targeting preferences. One of these interactions occurs between integrase and chromatin-associated host protein lens epithelium-derived growth factor (LEDGF)/p75 (241 – 243). Although this manuscript centers on CA-interacting proteins, we will briefly touch upon the role of LEDGF/p75 in HIV-1 integration targeting. When LEDGF/p75 was depleted from cells, preferences for the above-mentioned active chromatin annotations decreased across the board, though at the same time each of these targeting metrics remained enriched relative to random values (223, 242, 243). Under baseline infection conditions, HIV-1 integration favors gene mid-regions (244). In LEDGF/p75 knockout cells, this preference strikingly shifted to gene 5′ end regions, suggesting LEDGF/p75’s role in HIV-1 integration targeting may be linked to transcriptional elongation (223, 245). Most recently, LEDGF/p75 has been shown to play an important role in the targeting of TADs (237).
Initial investigations into the roles of CA-binding host factors revealed comparatively minor effects in HIV-1 integration targeting. Thus, while NUP358 deletion revealed significant reductions in the targeting of gene dense regions, integration into genes was reduced only slightly (246). Similarly, NUP153 depletion in one study was reported to significantly reduce integration into gene-dense regions without affecting the frequency of intragenic integration targeting (247). A separate study however revealed significant 8% and 5% reductions in intragenic targeting frequencies upon knockdown of NUP153 and NUP98, respectively (60). Curiously, infections conducted in the presence of cyclosporin A, which is an inhibitor of the capsid-CYPA interaction (112), increased integration into gene-dense regions without significantly affecting the percentage of intragenic integration targeting (52).
The second virus-host interaction observed to play a dominant role in HIV-1 integration targeting is CA-CPSF6. In CPSF6 knockout cells, integration into genes was reduced by as much as 25%, similar to the effect observed in LEDGF/p75 knockout cells. However, in contrast to the results observed in LEDGF/p75 knockout cells, other chromatin targeting metrics, namely gene-dense regions, activating epigenetic marks, and SPADs, became disfavored when CPSF6 was knocked out (223, 236). As a consequence, heterochromatin marks, such as repressive histone modifications and LADs, can become favored integration targets in CPSF6 knockout cells (202, 223). LEDGF/p75 and CPSF6 are lentiviral and primate lentiviral-specific host factors, respectively (58, 248 – 251), and the integration targeting profile of the γ-retrovirus Moloney murine leukemia virus was largely unchanged in LEDGF/p75 and CPSF6 knockout cells (223, 251). At the extent analyzed, proximity to SPADs and nuclear speckles (NS) is the strongest genomic predictors of HIV-1 integration targeting (252).
Recent image-based work has drawn interesting parallels between intranuclear localization of HIV-1 cores and integration targeting preferences. Viruses unable to interact with CPSF6 (through CA mutagenesis or CPSF6 knockdown/knockout) were arrested for nuclear penetration and uncharacteristically targeted LADs for integration (165, 168, 201 – 203) (Fig. 4A). Initial reports of roles for NUPs and CYPA in HIV-1 integration targeting may be interpreted in light of these more recent findings. NUP358 or NUP153 depletion may similarly restrict the extent of nuclear incursion of HIV-1 cores, leading to significant decreases in integration in gene-dense regions, which strongly track with SPADs (252). Conversely, because CYPA binding regulates the CA-CPSF6 interaction (107), loss of CYPA may simply increase CPSF6 occupancy, translating to further penetration distance than observed under baseline infection conditions. Such hypotheses can be tested by quantifying intranuclear penetration distances of HIV-1 cores under NUP and CYPA knockdown conditions.
Under baseline infection conditions, CPSF6, independent of cell type, relocalized from broadly pan-nuclear (nucleoli excluded) patterns to distinct puncta that co-localize with NS (175, 177, 203, 236, 253). This dramatic reorganization of nuclear CPSF6 strictly depended on its interaction with CA (177, 203, 236). Consistent with the notion that reverse transcription terminates in cell nuclei (176, 177, 236), viral DNA synthesis can occur in the context of CPSF6-HIV puncta (253). Imaging of live cells has indicated that fluorescently labeled CPSF6-HIV puncta are long-lived and highly dynamic (236, 240). Recovery of fluorescently labeled CPSF6 puncta within ~2 to 4 min of photobleaching was interpreted as evidence for liquid-liquid phase separation (LLPS) (240). LLPS of macromolecules drives intermolecular interactions that underlie the formation of biomolecular condensates and cellular bodies, including NS (254 – 256).
These observations have suggested the following model for the role of CPSF6 in HIV-1 nuclear trafficking and integration targeting (Fig. 4B). CPSF6 displaces the capsid from its interaction with NUP153 at the nuclear basket to license core penetration into the nucleoplasm (203). CPSF6 LLPS activity may then help to determine HIV-CPSF6 puncta formation and integration into SPAD chromosomal regions (240, 257), though this has yet to be formally demonstrated. Consistent with a potential role for LLPS activity, fluorescently tagged CPSF6 RSLD fusion proteins co-localize with NS in cells and display LLPS activity in vitro (125). Moreover, hyperosmotic stress can induce the formation of cellular CPSF6 foci, which is indicative of LLPS activity (258), and recombinant mCherry-CPSF6 fusion protein displayed LLPS activity in vitro (259). Several questions immediately arise from these observations. For example, might CPSF6 LLPS activty primarily underlie visible punta formation at NS, or perhaps also play a role to translocate the HIV-1 core through the nucleoplasm? CPSF6 contains two separate LCRs (the PrLD and RSLD) and although each LCR on its own can pase separate in vitro (125, 259), the contribution of each region to CPSF6’s overall LLPS activity needs to be established. Such observations raise the intriguing possibility that CPSF6 functionality beyond CA-binding might contribute to HIV-1 intranuclear trafficking and integration targeting. Additional questions include whether other capsid and/or CA-binding proteins play potential roles in CPSF6 puncta formation and HIV-1 integration targeting? Moving forward, we would propose that assays for CPSF6 puncta formation and SPAD-proximal integration should be incorporated into studies that examine the roles of CA-binding proteins in intranuclear trafficking and HIV-1 integration targeting.
CONCLUSIONS AND PERSPECTIVES
The HIV-1 ingress field has uncovered a dizzying array of human proteins that can interact with CA and the capsid (Table 1). Several questions arise based on these observations. What fraction of the interactions that play demonstrative roles in growth promotion are required for HIV-1 to successfully infect any given cell? Plausibly, some of these interactions may be cell-type specific, while others may be equally important regardless of cell type. Given the applicability of CRISPR-Cas9 with primary cells associated with HIV-1 research (260, 261), we would highlight the inclusion of knockdown/knockout experiments with primary cells moving forward. Because each capsid is constructed from ~200 (or so) hexamers and exactly 12 pentamers (Fig. 1C), there are several binding sites available at any given point during HIV-1 ingress. Thus, it is theoretically possible (and seems probable) that interactions with multiple host factors could occur at any given point in time. It would be informative if future studies could extend analyses of new capsid-host interactions to test importance alongside factor(s) that were previously established to play an important role at the indicated point of the infection cycle.
As cytoplasmic trafficking transits to nuclear translocation, new sets of CA-host interactions will occur. How cytoplasmic-specific interactions transfer to interactions specific to nuclear import is largely a black box and should be one focus of future work. For example, is it important for NUP358 to displace CYPA from the CYP-binding loop, or does NUP358 engage unoccupied CYP-binding loops to dock the core at the NPC? Similarly, does NUP153 need to displace SEC24C from FG-binding pockets, or might NUP153 and SEC24 simultaneously bind the capsid to form higher-order PrLD interactions along the mature lattice? There is reasonable evidence to suggest that CPSF6 displacement of NUP153 from capsid is important to transition from nuclear import to intranuclear trafficking (203). In the preceding section, we have highlighted several interesting questions that arise from the potential involvement of CPSF6 LLPS activity to intranuclear trafficking and HIV-1 integration targeting.
One of the most important aspects of basic scientific research is the potential to inform translational medicines. LEN is a highly potent capsid inhibitor that displays greater potency to inhibit HIV-1 ingress as compared to the late steps of virus replication (262). Based on efficient competition with FG host factors for capsid binding (262, 263), it has been proposed that inhibition of capsid-host interactions in part underlies LEN’s potency (263). However, the results of a more recent study have argued that inhibition of capsid-host interactions plays little if any role in determining the compound’s potency (122). Drug binding at the interface of two separate molecules, in this case two adjacent CA molecules that compose a single FG pocket within the hexamer, can elicit allosteric affects that may alter structure/function distal from the point of drug binding, in this case decreasing capsid pliability that may be required for uncoating and integration (122). This story is reminiscent of the allosteric integrase inhibitors (ALLINIs) that bind HIV-1 integrase at the LEDGF/p75 binding site, which is likewise composed of two separate viral protein molecules (264, 265). Inhibition of integrase-LEDGF/p75 binding was initially thought to underlie ALLINI potency (265). However, follow-up work showed that these compounds elicit uncontrolled integrase polymerization (266, 267), the consequences of which primarily inhibit HIV-1 particle morphogenesis in a LEDGF/p75-independent manner (268). Thus, for both LEN and ALLINIs, due to the engagement of viral pockets that are composed of more than one target molecule, antiviral potency is driven by allosteric affects felt throughout higher-order structures: the mature capsid lattice in one case, and aberrant linear and branched chain integrase polymers in another. These highly unpredictable antiviral mechanisms of action should inspire investigators to seek small molecule inhibitors of other virus-host interactions, especially in cases where more than one viral molecule composes the complete host factor binding site.
ACKNOWLEDGMENTS
This work was supported by US National Institutes of Health grants R37AI039394-27 and R01AI052014-20.
The authors declare no conflict of interest.
Contributor Information
Alan N. Engelman, Email: alan_engelman@dfci.harvard.edu.
Sebla Bulent Kutluay, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA .
REFERENCES
- 1. Paik J. 2022. Lenacapavir: first approval. Drugs 82:1499–1504. doi: 10.1007/s40265-022-01786-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Lerner G, Weaver N, Anokhin B, Spearman P. 2022. Advances in HIV-1 assembly. Viruses 14:478. doi: 10.3390/v14030478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Sundquist WI, Kräusslich H-G. 2012. HIV-1 assembly, budding, and maturation. Cold Spring Harb Perspect Med 2:a006924. doi: 10.1101/cshperspect.a006924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Qu K, Ke Z, Zila V, Anders-Össwein M, Glass B, Mücksch F, Müller R, Schultz C, Müller B, Kräusslich HG, Briggs JAG. 2021. Maturation of the matrix and viral membrane of HIV-1. Science 373:700–704. doi: 10.1126/science.abe6821 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Samal AB, Green TJ, Saad JS. 2022. Atomic view of the HIV-1 matrix lattice; implications on virus assembly and envelope incorporation. Proc Natl Acad Sci U S A 119:e2200794119. doi: 10.1073/pnas.2200794119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Kotov A, Zhou J, Flicker P, Aiken C. 1999. Association of Nef with the human immunodeficiency virus type 1 core. J Virol 73:8824–8830. doi: 10.1128/JVI.73.10.8824-8830.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Accola MA, Ohagen A, Göttlinger HG. 2000. Isolation of human immunodeficiency virus type 1 cores: retention of Vpr in the absence of p6. J Virol 74:6198–6202. doi: 10.1128/jvi.74.13.6198-6202.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Welker R, Hohenberg H, Tessmer U, Huckhagel C, Kräusslich HG. 2000. Biochemical and structural analysis of isolated mature cores of human immunodeficiency virus type 1. J Virol 74:1168–1177. doi: 10.1128/jvi.74.3.1168-1177.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Gres AT, Kirby KA, KewalRamani VN, Tanner JJ, Pornillos O, Sarafianos SG. 2015. X-ray crystal structures of native HIV-1 capsid protein reveal conformational variability. Science 349:99–103. doi: 10.1126/science.aaa5936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Zhao G, Perilla JR, Yufenyuy EL, Meng X, Chen B, Ning J, Ahn J, Gronenborn AM, Schulten K, Aiken C, Zhang P. 2013. Mature HIV-1 capsid structure by cryo-electron microscopy and all-atom molecular dynamics. Nature 497:643–646. doi: 10.1038/nature12162 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Price AJ, Jacques DA, McEwan WA, Fletcher AJ, Essig S, Chin JW, Halambage UD, Aiken C, James LC. 2014. Host cofactors and pharmacologic ligands share an essential interface in HIV-1 capsid that is lost upon disassembly. PLoS Pathog 10:e1004459. doi: 10.1371/journal.ppat.1004459 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Pornillos O, Ganser-Pornillos BK, Yeager M. 2011. Atomic-level modelling of the HIV capsid. Nature 469:424–427. doi: 10.1038/nature09640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Summers BJ, Digianantonio KM, Smaga SS, Huang PT, Zhou K, Gerber EE, Wang W, Xiong Y. 2019. Modular HIV-1 capsid assemblies reveal diverse host-capsid recognition mechanisms. Cell Host Microbe 26:203–216. doi: 10.1016/j.chom.2019.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Ansari-Lari MA, Donehower LA, Gibbs RA. 1995. Analysis of human immunodeficiency virus type 1 integrase mutants. Virology 213:332–335. [DOI] [PubMed] [Google Scholar]
- 15. Engelman A, Englund G, Orenstein JM, Martin MA, Craigie R. 1995. Multiple effects of mutations in human immunodeficiency virus type 1 Integrase on viral replication. J Virol 69:2729–2736. doi: 10.1128/JVI.69.5.2729-2736.1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Wiskerchen M, Muesing MA. 1995. Human immunodeficiency virus type 1 Integrase: effects of mutations on viral ability to integrate, direct viral gene expression from unintegrated viral DNA templates, and sustain viral propagation in primary cells. J Virol 69:376–386. doi: 10.1128/jvi.69.1.376-386.1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Wu Y, Marsh JW. 2001. Selective transcription and modulation of resting T cell activity by preintegrated HIV DNA. Science 293:1503–1506. doi: 10.1126/science.1061548 [DOI] [PubMed] [Google Scholar]
- 18. Wu Y, Marsh JW. 2003. Early transcription from nonintegrated DNA in human immunodeficiency virus infection. J Virol 77:10376–10382. doi: 10.1128/jvi.77.19.10376-10382.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Zhu Y, Wang GZ, Cingöz O, Goff SP. 2018. NP220 mediates silencing of unintegrated retroviral DNA. Nature 564:278–282. doi: 10.1038/s41586-018-0750-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Geis FK, Goff SP. 2019. Unintegrated HIV-1 DNAs are loaded with core and linker histones and transcriptionally silenced. Proc Natl Acad Sci U S A 116:23735–23742. doi: 10.1073/pnas.1912638116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Irwan ID, Karnowski HL, Bogerd HP, Tsai K, Cullen BR. 2020. Reversal of epigenetic silencing allows robust HIV-1 replication in the absence of integrase function. mBio 11:e01038-20. doi: 10.1128/mBio.01038-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Machida S, Depierre D, Chen HC, Thenin-Houssier S, Petitjean G, Doyen CM, Takaku M, Cuvier O, Benkirane M. 2020. Exploring histone loading on HIV DNA reveals a dynamic nucleosome positioning between unintegrated and integrated viral genome. Proc Natl Acad Sci U S A 117:6822–6830. doi: 10.1073/pnas.1913754117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Dupont L, Bloor S, Williamson JC, Cuesta SM, Shah R, Teixeira-Silva A, Naamati A, Greenwood EJD, Sarafianos SG, Matheson NJ, Lehner PJ. 2021. The SMC5/6 complex compacts and silences unintegrated HIV-1 DNA and is antagonized by Vpr. Cell Host Microbe 29:792–805. doi: 10.1016/j.chom.2021.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Geis FK, Sabo Y, Chen X, Li Y, Lu C, Goff SP. 2022. CHAF1A/B mediate silencing of unintegrated HIV-1 DNAs early in infection. Proc Natl Acad Sci U S A 119:e2116735119. doi: 10.1073/pnas.2116735119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Sakai H, Kawamura M, Sakuragi J, Sakuragi S, Shibata R, Ishimoto A, Ono N, Ueda S, Adachi A. 1993. Integration is essential for efficient gene expression of human immunodeficiency virus type 1. J Virol 67:1169–1174. doi: 10.1128/JVI.67.3.1169-1174.1993 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Englund G, Theodore TS, Freed EO, Engelman A, Martin MA. 1995. Integration is required for productive infection of monocyte-derived macrophages by human immunodeficiency virus type 1. J Virol 69:3216–3219. doi: 10.1128/jvi.69.5.3216-3219.1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Gao D, Wu J, Wu YT, Du F, Aroh C, Yan N, Sun L, Chen ZJ. 2013. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341:903–906. doi: 10.1126/science.1240933 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Lahaye X, Satoh T, Gentili M, Cerboni S, Conrad C, Hurbain I, El Marjou A, Lacabaratz C, Lelièvre JD, Manel N. 2013. The capsids of HIV-1 and HIV-2 determine immune detection of the viral cDNA by the innate sensor cGAS in dendritic cells. Immunity 39:1132–1142. doi: 10.1016/j.immuni.2013.11.002 [DOI] [PubMed] [Google Scholar]
- 29. Rasaiyaah J, Tan CP, Fletcher AJ, Price AJ, Blondeau C, Hilditch L, Jacques DA, Selwood DL, James LC, Noursadeghi M, Towers GJ. 2013. HIV-1 evades innate immune recognition through specific cofactor recruitment. Nature 503:402–405. doi: 10.1038/nature12769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Sumner RP, Harrison L, Touizer E, Peacock TP, Spencer M, Zuliani-Alvarez L, Towers GJ. 2020. Disrupting HIV-1 capsid formation causes cGAS sensing of viral DNA. EMBO J 39:e103958. doi: 10.15252/embj.2019103958 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Yoh SM, Mamede JI, Lau D, Ahn N, Sánchez-Aparicio MT, Temple J, Tuckwell A, Fuchs NV, Cianci GC, Riva L, Curry H, Yin X, Gambut S, Simons LM, Hultquist JF, König R, Xiong Y, García-Sastre A, Böcking T, Hope TJ, Chanda SK. 2022. Recognition of HIV-1 capsid by PQBP1 licenses an innate immune sensing of nascent HIV-1 DNA. Mol Cell 82:2871–2884. doi: 10.1016/j.molcel.2022.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Papa G, Albecka A, Mallery D, Vaysburd M, Renner N, James LC. 2023. IP6-stabilised HIV capsids evade cGAS/STING-mediated host immune sensing. EMBO Rep 24:e56275. doi: 10.15252/embr.202256275 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Lahaye X, Gentili M, Silvin A, Conrad C, Picard L, Jouve M, Zueva E, Maurin M, Nadalin F, Knott GJ, Zhao B, Du F, Rio M, Amiel J, Fox AH, Li P, Etienne L, Bond CS, Colleaux L, Manel N. 2018. NONO detects the nuclear HIV capsid to promote cGAS-mediated innate immune activation. Cell 175:488–501. doi: 10.1016/j.cell.2018.08.062 [DOI] [PubMed] [Google Scholar]
- 34. Temple J, Tripler TN, Shen Q, Xiong Y. 2020. A snapshot of HIV-1 capsid-host interactions. Curr Res Struct Biol 2:222–228. doi: 10.1016/j.crstbi.2020.10.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. AlBurtamani N, Paul A, Fassati A. 2021. The role of capsid in the early steps of HIV-1 infection: new insights into the core of the matter. Viruses 13:1161. doi: 10.3390/v13061161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Engelman AN. 2021. HIV capsid and integration targeting. Viruses 13:125. doi: 10.3390/v13010125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Naghavi MH. 2021. HIV-1 capsid exploitation of the host microtubule cytoskeleton during early infection. Retrovirology 18:19. doi: 10.1186/s12977-021-00563-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Rossi E, Meuser ME, Cunanan CJ, Cocklin S. 2021. Structure, function, and interactions of the HIV-1 capsid protein. Life (Basel) 11:100. doi: 10.3390/life11020100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Scoca V, Di Nunzio F. 2021. The HIV-1 capsid: from structural component to key factor for host nuclear invasion. Viruses 13:273. doi: 10.3390/v13020273 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Shen Q, Wu C, Freniere C, Tripler TN, Xiong Y. 2021. Nuclear import of HIV-1. Viruses 13:2242. doi: 10.3390/v13112242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Toccafondi E, Lener D, Negroni M. 2021. HIV-1 capsid core: a bullet to the heart of the target cell. Front Microbiol 12:652486. doi: 10.3389/fmicb.2021.652486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Wang W, Li Y, Zhang Z, Wei W. 2022. Human immunodeficiency virus-1 core: the Trojan horse in virus-host interaction. Front Microbiol 13:1002476. doi: 10.3389/fmicb.2022.1002476 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Cáceres JF, Misteli T, Screaton GR, Spector DL, Krainer AR. 1997. Role of the modular domains of SR proteins in subnuclear localization and alternative splicing specificity. J Cell Biol 138:225–238. doi: 10.1083/jcb.138.2.225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Lai MC, Lin RI, Tarn WY. 2001. Transportin-SR2 mediates nuclear import of phosphorylated SR proteins. Proc Natl Acad Sci U S A 98:10154–10159. doi: 10.1073/pnas.181354098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Gitti RK, Lee BM, Walker J, Summers MF, Yoo S, Sundquist WI. 1996. Structure of the amino-terminal core domain of the HIV-1 capsid protein. Science 273:231–235. doi: 10.1126/science.273.5272.231 [DOI] [PubMed] [Google Scholar]
- 46. Gamble TR, Yoo S, Vajdos FF, von Schwedler UK, Worthylake DK, Wang H, McCutcheon JP, Sundquist WI, Hill CP. 1997. Structure of the carboxyl-terminal dimerization domain of the HIV-1 capsid protein. Science 278:849–853. doi: 10.1126/science.278.5339.849 [DOI] [PubMed] [Google Scholar]
- 47. Pornillos O, Ganser-Pornillos BK, Kelly BN, Hua Y, Whitby FG, Stout CD, Sundquist WI, Hill CP, Yeager M. 2009. X-ray structures of the hexameric building block of the HIV capsid. Cell 137:1282–1292. doi: 10.1016/j.cell.2009.04.063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Schirra RT, Dos Santos NFB, Zadrozny KK, Kucharska I, Ganser-Pornillos BK, Pornillos O. 2023. A molecular switch modulates assembly and host factor binding of the HIV-1 capsid. Nat Struct Mol Biol 30:383–390. doi: 10.1038/s41594-022-00913-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Ganser BK, Li S, Klishko VY, Finch JT, Sundquist WI. 1999. Assembly and analysis of conical models for the HIV-1 core. Science 283:80–83. doi: 10.1126/science.283.5398.80 [DOI] [PubMed] [Google Scholar]
- 50. Mattei S, Glass B, Hagen WJH, Kräusslich H-G, Briggs JAG. 2016. The structure and flexibility of conical HIV-1 capsids determined within intact virions. Science 354:1434–1437. doi: 10.1126/science.aah4972 [DOI] [PubMed] [Google Scholar]
- 51. Gamble TR, Vajdos FF, Yoo S, Worthylake DK, Houseweart M, Sundquist WI, Hill CP. 1996. Crystal structure of human cyclophilin A bound to the amino-terminal domain of HIV-1 capsid. Cell 87:1285–1294. doi: 10.1016/s0092-8674(00)81823-1 [DOI] [PubMed] [Google Scholar]
- 52. Schaller T, Ocwieja KE, Rasaiyaah J, Price AJ, Brady TL, Roth SL, Hué S, Fletcher AJ, Lee K, KewalRamani VN, Noursadeghi M, Jenner RG, James LC, Bushman FD, Towers GJ. 2011. HIV-1 capsid-cyclophilin interactions determine nuclear import pathway, integration targeting and replication efficiency. PLoS Pathog 7:e1002439. doi: 10.1371/journal.ppat.1002439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Matreyek KA, Yücel SS, Li X, Engelman A. 2013. Nucleoporin NUP153 phenylalanine-glycine motifs engage a common binding pocket within the HIV-1 capsid protein to mediate lentiviral infectivity. PLoS Pathog 9:e1003693. doi: 10.1371/journal.ppat.1003693 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Bhattacharya A, Alam SL, Fricke T, Zadrozny K, Sedzicki J, Taylor AB, Demeler B, Pornillos O, Ganser-Pornillos BK, Diaz-Griffero F, Ivanov DN, Yeager M. 2014. Structural basis of HIV-1 capsid recognition by PF74 and CPSF6. Proc Natl Acad Sci U S A 111:18625–18630. doi: 10.1073/pnas.1419945112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Li S, Hill CP, Sundquist WI, Finch JT. 2000. Image reconstructions of helical assemblies of the HIV-1 CA protein. Nature 407:409–413. doi: 10.1038/35030177 [DOI] [PubMed] [Google Scholar]
- 56. Dick RA, Zadrozny KK, Xu C, Schur FKM, Lyddon TD, Ricana CL, Wagner JM, Perilla JR, Ganser-Pornillos BK, Johnson MC, Pornillos O, Vogt VM. 2018. Inositol phosphates are assembly co-factors for HIV-1. Nature 560:509–512. doi: 10.1038/s41586-018-0396-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Stremlau M, Perron M, Lee M, Li Y, Song B, Javanbakht H, Diaz-Griffero F, Anderson DJ, Sundquist WI, Sodroski J. 2006. Specific recognition and accelerated uncoating of retroviral capsids by the TRIM5α restriction factor. Proc Natl Acad Sci U S A 103:5514–5519. doi: 10.1073/pnas.0509996103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Lee K, Ambrose Z, Martin TD, Oztop I, Mulky A, Julias JG, Vandegraaff N, Baumann JG, Wang R, Yuen W, Takemura T, Shelton K, Taniuchi I, Li Y, Sodroski J, Littman DR, Coffin JM, Hughes SH, Unutmaz D, Engelman A, KewalRamani VN. 2010. Flexible use of nuclear import pathways by HIV-1. Cell Host Microbe 7:221–233. doi: 10.1016/j.chom.2010.02.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Di Nunzio F, Danckaert A, Fricke T, Perez P, Fernandez J, Perret E, Roux P, Shorte S, Charneau P, Diaz-Griffero F, Arhel NJ. 2012. Human nucleoporins promote HIV-1 docking at the nuclear pore, nuclear import and integration. PLoS One 7:e46037. doi: 10.1371/journal.pone.0046037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Di Nunzio F, Fricke T, Miccio A, Valle-Casuso JC, Perez P, Souque P, Rizzi E, Severgnini M, Mavilio F, Charneau P, Diaz-Griffero F. 2013. Nup153 and Nup98 bind the HIV-1 core and contribute to the early steps of HIV-1 replication. Virology 440:8–18. doi: 10.1016/j.virol.2013.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Fribourgh JL, Nguyen HC, Matreyek KA, Alvarez FJD, Summers BJ, Dewdney TG, Aiken C, Zhang P, Engelman A, Xiong Y. 2014. Structural insight into HIV-1 restriction by MxB. Cell Host Microbe 16:627–638. doi: 10.1016/j.chom.2014.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Fricke T, White TE, Schulte B, de Souza Aranha Vieira DA, Dharan A, Campbell EM, Brandariz-Nuñez A, Diaz-Griffero F. 2014. MxB binds to the HIV-1 core and prevents the uncoating process of HIV-1. Retrovirology 11:68. doi: 10.1186/s12977-014-0068-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Selyutina A, Bulnes-Ramos A, Diaz-Griffero F. 2018. Binding of host factors to stabilized HIV-1 capsid tubes. Virology 523:1–5. doi: 10.1016/j.virol.2018.07.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Highland CM, Tan A, Ricaña CL, Briggs JAG, Dick RA. 2023. Structural insights into HIV-1 polyanion-dependent capsid lattice formation revealed by single particle cryo-EM. Proc Natl Acad Sci U S A 120:e2220545120. doi: 10.1073/pnas.2220545120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Ni T, Zhu Y, Yang Z, Xu C, Chaban Y, Nesterova T, Ning J, Böcking T, Parker MW, Monnie C, Ahn J, Perilla JR, Zhang P. 2021. Structure of native HIV-1 cores and their interactions with IP6 and CypA. Sci Adv 7:eabj5715. doi: 10.1126/sciadv.abj5715 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Dharan A, Opp S, Abdel-Rahim O, Keceli SK, Imam S, Diaz-Griffero F, Campbell EM. 2017. Bicaudal D2 facilitates the cytoplasmic trafficking and nuclear import of HIV-1 genomes during infection. Proc Natl Acad Sci U S A 114:E10707–E10716. doi: 10.1073/pnas.1712033114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Carnes SK, Zhou J, Aiken C, Sundquist WI. 2018. HIV-1 engages a dynein-dynactin-BICD2 complex for infection and transport to the nucleus. J Virol 92:e00358-18. doi: 10.1128/JVI.00358-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Mitra S, Shanmugapriya S, Santos da Silva E, Naghavi MH, Kirchhoff F. 2020. HIV-1 exploits CLASP2 to induce microtubule stabilization and facilitate virus trafficking to the nucleus. J Virol 94:e00404-20. doi: 10.1128/JVI.00404-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Santos da Silva E, Shanmugapriya S, Malikov V, Gu F, Delaney MK, Naghavi MH. 2020. HIV-1 capsids mimic a microtubule regulator to coordinate early stages of infection. EMBO J 39:e104870. doi: 10.15252/embj.2020104870 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Price AJ, Fletcher AJ, Schaller T, Elliott T, Lee K, KewalRamani VN, Chin JW, Towers GJ, James LC, Krausslich H-G. 2012. CPSF6 defines a conserved capsid interface that modulates HIV-1 replication. PLoS Pathog 8:e1002896. doi: 10.1371/journal.ppat.1002896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Luban J, Bossolt KL, Franke EK, Kalpana GV, Goff SP. 1993. Human immunodeficiency virus type 1 Gag protein binds to cyclophilins A and B. Cell 73:1067–1078. doi: 10.1016/0092-8674(93)90637-6 [DOI] [PubMed] [Google Scholar]
- 72. Liu C, Perilla JR, Ning J, Lu M, Hou G, Ramalho R, Himes BA, Zhao G, Bedwell GJ, Byeon I-J, Ahn J, Gronenborn AM, Prevelige PE, Rousso I, Aiken C, Polenova T, Schulten K, Zhang P. 2016. Cyclophilin A stabilizes the HIV-1 capsid through a novel non-canonical binding site. Nat Commun 7:10714. doi: 10.1038/ncomms10714 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Ni T, Gerard S, Zhao G, Dent K, Ning J, Zhou J, Shi J, Anderson-Daniels J, Li W, Jang S, Engelman AN, Aiken C, Zhang P. 2020. Intrinsic curvature of the HIV-1 CA hexamer underlies capsid topology and interaction with cyclophilin A. Nat Struct Mol Biol 27:855–862. doi: 10.1038/s41594-020-0467-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. DeBoer J, Madson CJ, Belshan M. 2016. Cyclophilin B enhances HIV-1 infection. Virology 489:282–291. doi: 10.1016/j.virol.2015.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Maillet S, Fernandez J, Decourcelle M, El Koulali K, Blanchet FP, Arhel NJ, Maarifi G, Nisole S. 2020. Daxx inhibits HIV-1 reverse transcription and uncoating in a SUMO-dependent manner. Viruses 12:636. doi: 10.3390/v12060636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Shanmugapriya S, Santos da Silva E, Campbell JA, Boisjoli M-P, Naghavi MH. 2021. Dynactin 1 negatively regulates HIV-1 infection by sequestering the host cofactor CLIP170. Proc Natl Acad Sci U S A 118:e2102884118. doi: 10.1073/pnas.2102884118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Delaney MK, Malikov V, Chai Q, Zhao G, Naghavi MH. 2017. Distinct functions of diaphanous-related formins regulate HIV-1 uncoating and transport. Proc Natl Acad Sci U S A 114:E6932–E6941. doi: 10.1073/pnas.1700247114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Malikov V, da Silva ES, Jovasevic V, Bennett G, de Souza Aranha Vieira DA, Schulte B, Diaz-Griffero F, Walsh D, Naghavi MH. 2015. HIV-1 capsids bind and exploit the kinesin-1 adaptor FEZ1 for inward movement to the nucleus. Nat Commun 6:6660. doi: 10.1038/ncomms7660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Huang P-T, Summers BJ, Xu C, Perilla JR, Malikov V, Naghavi MH, Xiong Y. 2019. FEZ1 is recruited to a conserved cofactor site on capsid to promote HIV-1 trafficking. Cell Rep 28:2373–2385. doi: 10.1016/j.celrep.2019.07.079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Fernandez J, Portilho DM, Danckaert A, Munier S, Becker A, Roux P, Zambo A, Shorte S, Jacob Y, Vidalain PO, Charneau P, Clavel F, Arhel NJ. 2015. Microtubule-associated proteins 1 (MAP1) promote human immunodeficiency virus type I (HIV-1) intracytoplasmic routing to the nucleus. J Biol Chem 290:4631–4646. doi: 10.1074/jbc.M114.613133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Dochi T, Nakano T, Inoue M, Takamune N, Shoji S, Sano K, Misumi S. 2014. Phosphorylation of human immunodeficiency virus type 1 capsid protein at serine 16, required for peptidyl-prolyl isomerase-dependent uncoating, is mediated by virion-incorporated extracellular signal-regulated kinase 2. J Gen Virol 95:1156–1166. doi: 10.1099/vir.0.060053-0 [DOI] [PubMed] [Google Scholar]
- 82. Malikov V, Naghavi MH. 2017. Localized phosphorylation of a kinesin-1 adaptor by a capsid-associated kinase regulates HIV-1 motility and uncoating. Cell Rep 20:2792–2799. doi: 10.1016/j.celrep.2017.08.076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Takeuchi H, Saito H, Noda T, Miyamoto T, Yoshinaga T, Terahara K, Ishii H, Tsunetsugu-Yokota Y, Yamaoka S, Emerman M. 2017. Phosphorylation of the HIV-1 capsid by MELK triggers uncoating to promote viral cDNA synthesis. PLoS Pathog 13:e1006441. doi: 10.1371/journal.ppat.1006441 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kong J, Xu B, Wei W, Wang X, Xie W, Yu XF. 2014. Characterization of the amino-terminal domain of Mx2/MxB-dependent interaction with the HIV-1 capsid. Protein Cell 5:954–957. doi: 10.1007/s13238-014-0113-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Buffone C, Schulte B, Opp S, Diaz-Griffero F, Ross SR. 2015. Contribution of MxB oligomerization to HIV-1 capsid binding and restriction. J Virol 89:3285–3294. doi: 10.1128/JVI.03730-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Betancor G, Dicks MDJ, Jimenez-Guardeño JM, Ali NH, Apolonia L, Malim MH. 2019. The GTPase domain of MX2 interacts with the HIV-1 capsid, enabling its short isoform to moderate antiviral restriction. Cell Rep 29:1923–1933. doi: 10.1016/j.celrep.2019.10.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Schulte B, Buffone C, Opp S, Di Nunzio F, De Souza Aranha Vieira DA, Brandariz-Nuñez A, Diaz-Griffero F. 2015. Restriction of HIV-1 requires the N-terminal region of MxB as a capsid-binding motif but not as a nuclear localization signal. J Virol 89:8599–8610. doi: 10.1128/JVI.00753-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Buffone C, Martinez-Lopez A, Fricke T, Opp S, Severgnini M, Cifola I, Petiti L, Frabetti S, Skorupka K, Zadrozny KK, Ganser-Pornillos BK, Pornillos O, Di Nunzio F, Diaz-Griffero F. 2018. Nup153 unlocks the nuclear pore complex for HIV-1 nuclear translocation in nondividing cells. J Virol 92:e00648-18. doi: 10.1128/JVI.00648-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Shen Q, Kumari S, Xu C, Jang S, Shi J, Burdick RC, Levintov L, Xiong Q, Wu C, Devarkar SC, Tian T, Tripler TN, Hu Y, Yuan S, Temple J, Feng Q, Lusk CP, Aiken C, Engelman AN, Perilla JR, Pathak VK, Lin C, Xiong Y. 2023. The capsid lattice engages a bipartite NUP153 motif to mediate nuclear entry of HIV-1 cores. Proc Natl Acad Sci U S A 120:e2202815120. doi: 10.1073/pnas.2202815120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Rebensburg SV, Wei G, Larue RC, Lindenberger J, Francis AC, Annamalai AS, Morrison J, Shkriabai N, Huang S-W, KewalRamani V, Poeschla EM, Melikyan GB, Kvaratskhelia M. 2021. Sec24C is an HIV-1 host dependency factor crucial for virus replication. Nat Microbiol 6:435–444. doi: 10.1038/s41564-021-00868-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Bichel K, Price AJ, Schaller T, Towers GJ, Freund SMV, James LC. 2013. HIV-1 capsid undergoes coupled binding and isomerization by the nuclear pore protein NUP358. Retrovirology 10:81. doi: 10.1186/1742-4690-10-81 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Lin DH, Zimmermann S, Stuwe T, Stuwe E, Hoelz A. 2013. Structural and functional analysis of the C-terminal domain of Nup358/RanBP2. J Mol Biol 425:1318–1329. doi: 10.1016/j.jmb.2013.01.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Kane M, Rebensburg SV, Takata MA, Zang TM, Yamashita M, Kvaratskhelia M, Bieniasz PD. 2018. Nuclear pore heterogeneity influences HIV-1 infection and the antiviral activity of MX2. Elife 7:e35738. doi: 10.7554/eLife.35738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Shen Q, Feng Q, Wu C, Xiong Q, Tian T, Yuan S, Shi J, Bedwell GJ, Yang R, Aiken C, Engelman AN, Lusk CP, Lin C, Xiong Y. 2023. Modeling HIV-1 nuclear entry with nucleoporin-gated DNA-origami channels. Nat Struct Mol Biol 30:425–435. doi: 10.1038/s41594-023-00925-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Guth CA, Sodroski J. 2014. Contribution of PDZD8 to stabilization of the human immunodeficiency virus type 1 capsid. J Virol 88:4612–4623. doi: 10.1128/JVI.02945-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Misumi S, Inoue M, Dochi T, Kishimoto N, Hasegawa N, Takamune N, Shoji S. 2010. Uncoating of human immunodeficiency virus type 1 requires prolyl isomerase Pin1. J Biol Chem 285:25185–25195. doi: 10.1074/jbc.M110.114256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Schaller T, Bulli L, Pollpeter D, Betancor G, Kutzner J, Apolonia L, Herold N, Burk R, Malim MH. 2017. Effects of inner nuclear membrane proteins SUN1/UNC-84A and SUN2/UNC-84B on the early steps of HIV-1 infection. J Virol 91:e00463-17. doi: 10.1128/JVI.00463-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Persaud M, Selyutina A, Buffone C, Opp S, Donahue DA, Schwartz O, Diaz-Griffero F. 2021. Nuclear restriction of HIV-1 infection by SUN1. Sci Rep 11:19128. doi: 10.1038/s41598-021-98541-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Fernandez J, Machado AK, Lyonnais S, Chamontin C, Gärtner K, Léger T, Henriquet C, Garcia C, Portilho DM, Pugnière M, Chaloin L, Muriaux D, Yamauchi Y, Blaise M, Nisole S, Arhel NJ. 2019. Transportin-1 binds to the HIV-1 capsid via a nuclear localization signal and triggers uncoating. Nat Microbiol 4:1840–1850. doi: 10.1038/s41564-019-0575-6 [DOI] [PubMed] [Google Scholar]
- 100. Valle-Casuso JC, Di Nunzio F, Yang Y, Reszka N, Lienlaf M, Arhel N, Perez P, Brass AL, Diaz-Griffero F. 2012. TNPO3 is required for HIV-1 replication after nuclear import but prior to integration and binds the HIV-1 core. J Virol 86:5931–5936. doi: 10.1128/JVI.00451-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Yuan T, Yao W, Tokunaga K, Yang R, Sun B. 2016. An HIV-1 capsid binding protein TRIM11 accelerates viral uncoating. Retrovirology 13:72. doi: 10.1186/s12977-016-0306-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Ohainle M, Kim K, Komurlu Keceli S, Felton A, Campbell E, Luban J, Emerman M, Evans DT. 2020. TRIM34 restricts HIV-1 and SIV capsids in a TRIM5α-dependent manner. PLoS Pathog 16:e1008507. doi: 10.1371/journal.ppat.1008507 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Ganser-Pornillos BK, Chandrasekaran V, Pornillos O, Sodroski JG, Sundquist WI, Yeager M. 2011. Hexagonal assembly of a restricting TRIM5α protein. Proc Natl Acad Sci U S A 108:534–539. doi: 10.1073/pnas.1013426108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Skorupka KA, Roganowicz MD, Christensen DE, Wan Y, Pornillos O, Ganser-Pornillos BK. 2019. Hierarchical assembly governs TRIM5α recognition of HIV-1 and retroviral capsids. Sci Adv 5:eaaw3631. doi: 10.1126/sciadv.aaw3631 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Sebastian S, Luban J. 2005. TRIM5α selectively binds a restriction-sensitive retroviral capsid. Retrovirology 2:40. doi: 10.1186/1742-4690-2-40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Thul PJ, Åkesson L, Wiking M, Mahdessian D, Geladaki A, Ait Blal H, Alm T, Asplund A, Björk L, Breckels LM, Bäckström A, Danielsson F, Fagerberg L, Fall J, Gatto L, Gnann C, Hober S, Hjelmare M, Johansson F, Lee S, Lindskog C, Mulder J, Mulvey CM, Nilsson P, Oksvold P, Rockberg J, Schutten R, Schwenk JM, Sivertsson Å, Sjöstedt E, Skogs M, Stadler C, Sullivan DP, Tegel H, Winsnes C, Zhang C, Zwahlen M, Mardinoglu A, Pontén F, von Feilitzen K, Lilley KS, Uhlén M, Lundberg E. 2017. A subcellular map of the human proteome. Science 356:eaal3321. doi: 10.1126/science.aal3321 [DOI] [PubMed] [Google Scholar]
- 107. Zhong Z, Ning J, Boggs EA, Jang S, Wallace C, Telmer C, Bruchez MP, Ahn J, Engelman AN, Zhang P, Watkins SC, Ambrose Z, Liu S-L, Smithgall TE. 2021. Cytoplasmic CPSF6 regulates HIV-1 capsid trafficking and infection in a cyclophilin A-dependent manner. mBio 12:e03142-20. doi: 10.1128/mBio.03142-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Pontén F, Jirström K, Uhlen M. 2008. The Human protein atlas--a tool for pathology. J Pathol 216:387–393. doi: 10.1002/path.2440 [DOI] [PubMed] [Google Scholar]
- 109. Chartrain I, Blot J, Lerivray H, Guyot N, Tassan JP. 2007. A mitochondrial-targeting signal is present in the non-catalytic domain of the MELK protein kinase. Cell Biol Int 31:196–201. doi: 10.1016/j.cellbi.2006.10.005 [DOI] [PubMed] [Google Scholar]
- 110. Goujon C, Moncorgé O, Bauby H, Doyle T, Barclay WS, Malim MH, Doms RW. 2014. Transfer of the amino-terminal nuclear envelope targeting domain of human MX2 converts MX1 into an HIV-1 resistance factor. J Virol 88:9017–9026. doi: 10.1128/JVI.01269-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Franke EK, Yuan HEH, Luban J. 1994. Specific incorporation of cyclophilin A into HIV-1 virions. Nature 372:359–362. doi: 10.1038/372359a0 [DOI] [PubMed] [Google Scholar]
- 112. Thali M, Bukovsky A, Kondo E, Rosenwirth B, Walsh CT, Sodroski J, Göttlinger HG. 1994. Functional association of cyclophilin A with HIV-1 virions. Nature 372:363–365. doi: 10.1038/372363a0 [DOI] [PubMed] [Google Scholar]
- 113. Sokolskaja E, Sayah DM, Luban J. 2004. Target cell cyclophilin A modulates human immunodeficiency virus type 1 infectivity. J Virol 78:12800–12808. doi: 10.1128/JVI.78.23.12800-12808.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Hatziioannou T, Perez-Caballero D, Cowan S, Bieniasz PD. 2005. Cyclophilin interactions with incoming human immunodeficiency virus type 1 capsids with opposing effects on infectivity in human cells. J Virol 79:176–183. doi: 10.1128/JVI.79.1.176-183.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. De Iaco A, Luban J. 2014. Cyclophilin A promotes HIV-1 reverse transcription but its effect on transduction correlates best with its effect on nuclear entry of viral cDNA. Retrovirology 11:11. doi: 10.1186/1742-4690-11-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Luo X, Yang W, Gao G. 2018. SUN1 regulates HIV-1 nuclear import in a manner dependent on the interaction between the viral capsid and cellular cyclophilin A. J Virol 92:e00229-18. doi: 10.1128/JVI.00229-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Lahaye X, Satoh T, Gentili M, Cerboni S, Silvin A, Conrad C, Ahmed-Belkacem A, Rodriguez EC, Guichou JF, Bosquet N, Piel M, Le Grand R, King MC, Pawlotsky JM, Manel N. 2016. Nuclear envelope protein SUN2 promotes cyclophilin-A-dependent steps of HIV replication. Cell Rep 15:879–892. doi: 10.1016/j.celrep.2016.03.074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Kim K, Dauphin A, Komurlu S, McCauley SM, Yurkovetskiy L, Carbone C, Diehl WE, Strambio-De-Castillia C, Campbell EM, Luban J. 2019. Cyclophilin A protects HIV-1 from restriction by human TRIM5α. Nat Microbiol 4:2044–2051. doi: 10.1038/s41564-019-0592-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Selyutina A, Persaud M, Simons LM, Bulnes-Ramos A, Buffone C, Martinez-Lopez A, Scoca V, Di Nunzio F, Hiatt J, Marson A, Krogan NJ, Hultquist JF, Diaz-Griffero F. 2020. Cyclophilin A prevents HIV-1 restriction in lymphocytes by blocking human TRIM5α binding to the viral core. Cell Rep 30:3766–3777. doi: 10.1016/j.celrep.2020.02.100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Miles RJ, Kerridge C, Hilditch L, Monit C, Jacques DA, Towers GJ. 2020. MxB sensitivity of HIV-1 is determined by a highly variable and dynamic capsid surface. Elife 9:e56910. doi: 10.7554/eLife.56910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Braaten D, Luban J. 2001. Cyclophilin A regulates HIV-1 infectivity, as demonstrated by gene targeting in human T cells. EMBO J 20:1300–1309. doi: 10.1093/emboj/20.6.1300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Wei G, Iqbal N, Courouble VV, Francis AC, Singh PK, Hudait A, Annamalai AS, Bester S, Huang S-W, Shkriabai N, Briganti L, Haney R, KewalRamani VN, Voth GA, Engelman AN, Melikyan GB, Griffin PR, Asturias F, Kvaratskhelia M. 2022. Prion-like low complexity regions enable avid virus-host interactions during HIV-1 infection. Nat Commun 13:5879. doi: 10.1038/s41467-022-33662-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Stacey JCV, Tan A, Lu JM, James LC, Dick RA, Briggs JAG. 2023. Two structural switches in HIV-1 capsid regulate capsid curvature and host factor binding. Proc Natl Acad Sci U S A 120:e2220557120. doi: 10.1073/pnas.2220557120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Lancaster AK, Nutter-Upham A, Lindquist S, King OD. 2014. PLAAC: a web and command-line application to identify proteins with prion-like amino acid composition. Bioinformatics 30:2501–2502. doi: 10.1093/bioinformatics/btu310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Greig JA, Nguyen TA, Lee M, Holehouse AS, Posey AE, Pappu RV, Jedd G. 2020. Arginine-enriched mixed-charge domains provide cohesion for nuclear speckle condensation. Mol Cell 77:1237–1250. doi: 10.1016/j.molcel.2020.01.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Jacques DA, McEwan WA, Hilditch L, Price AJ, Towers GJ, James LC. 2016. HIV-1 uses dynamic capsid pores to import nucleotides and fuel encapsidated DNA synthesis. Nature 536:349–353. doi: 10.1038/nature19098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Xu C, Fischer DK, Rankovic S, Li W, Dick RA, Runge B, Zadorozhnyi R, Ahn J, Aiken C, Polenova T, Engelman AN, Ambrose Z, Rousso I, Perilla JR, Campbell EM. 2020. Permeability of the HIV-1 capsid to metabolites modulates viral DNA synthesis. PLoS Biol 18:e3001015. doi: 10.1371/journal.pbio.3001015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Mallery DL, Márquez CL, McEwan WA, Dickson CF, Jacques DA, Anandapadamanaban M, Bichel K, Towers GJ, Saiardi A, Böcking T, James LC. 2018. IP6 is an HIV pocket factor that prevents capsid collapse and promotes DNA synthesis. Elife 7:e35335. doi: 10.7554/eLife.35335 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Mallery DL, Kleinpeter AB, Renner N, Faysal KMR, Novikova M, Kiss L, Wilson MSC, Ahsan B, Ke Z, Briggs JAG, Saiardi A, Böcking T, Freed EO, James LC. 2021. A stable immature lattice packages IP(6) for HIV capsid maturation. Sci Adv 7:eabe4716. doi: 10.1126/sciadv.abe4716 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Renner N, Kleinpeter A, Mallery DL, Albecka A, Rifat Faysal KM, Böcking T, Saiardi A, Freed EO, James LC. 2023. HIV-1 is dependent on its immature lattice to recruit IP6 for mature capsid assembly. Nat Struct Mol Biol 30:370–382. doi: 10.1038/s41594-022-00887-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Gupta M, Pak AJ, Voth GA. 2023. Critical mechanistic features of HIV-1 viral capsid assembly. Sci Adv 9:eadd7434. doi: 10.1126/sciadv.add7434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Christensen DE, Ganser-Pornillos BK, Johnson JS, Pornillos O, Sundquist WI. 2020. Reconstitution and visualization of HIV-1 capsid-dependent replication and integration in vitro. Science 370:eabc8420. doi: 10.1126/science.abc8420 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Jennings J, Shi J, Varadarajan J, Jamieson PJ, Aiken C, Smithgall TE. 2020. The host cell metabolite inositol hexakisphosphate promotes efficient endogenous HIV-1 reverse transcription by stabilizing the viral capsid. mBio 11:e02820-20. doi: 10.1128/mBio.02820-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Yoh SM, Schneider M, Seifried J, Soonthornvacharin S, Akleh RE, Olivieri KC, De Jesus PD, Ruan C, de Castro E, Ruiz PA, Germanaud D, des Portes V, García-Sastre A, König R, Chanda SK. 2015. PQBP1 is a proximal sensor of the cGAS-dependent innate response to HIV-1. Cell 161:1293–1305. doi: 10.1016/j.cell.2015.04.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Chintala K, Mohareer K, Banerjee S. 2021. Dodging the host interferon-stimulated gene mediated innate immunity by HIV-1: a brief update on intrinsic mechanisms and counter-mechanisms. Front Immunol 12:716927. doi: 10.3389/fimmu.2021.716927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Hadpech S, Moonmuang S, Chupradit K, Yasamut U, Tayapiwatana C. 2022. Updating on roles of HIV intrinsic factors: a review of their antiviral mechanisms and emerging functions. Intervirology 65:67–79. doi: 10.1159/000519241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Stremlau M, Owens CM, Perron MJ, Kiessling M, Autissier P, Sodroski J. 2004. The cytoplasmic body component TRIM5α restricts HIV-1 infection in Old World monkeys. Nature 427:848–853. doi: 10.1038/nature02343 [DOI] [PubMed] [Google Scholar]
- 138. Dutrieux J, Maarifi G, Portilho DM, Arhel NJ, Chelbi-Alix MK, Nisole S, Aiken C. 2015. PML/TRIM19-dependent inhibition of retroviral reverse-transcription by Daxx. PLoS Pathog 11:e1005280. doi: 10.1371/journal.ppat.1005280 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Goujon C, Moncorgé O, Bauby H, Doyle T, Ward CC, Schaller T, Hué S, Barclay WS, Schulz R, Malim MH. 2013. Human MX2 is an interferon-induced post-entry inhibitor of HIV-1 infection. Nature 502:559–562. doi: 10.1038/nature12542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Kane M, Yadav SS, Bitzegeio J, Kutluay SB, Zang T, Wilson SJ, Schoggins JW, Rice CM, Yamashita M, Hatziioannou T, Bieniasz PD. 2013. MX2 is an interferon-induced inhibitor of HIV-1 infection. Nature 502:563–566. doi: 10.1038/nature12653 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Matreyek KA, Wang W, Serrao E, Singh PK, Levin HL, Engelman A. 2014. Host and viral determinants for MxB restriction of HIV-1 infection. Retrovirology 11:90. doi: 10.1186/s12977-014-0090-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Staeheli P, Haller O. 2018. Human Mx2/MxB: a potent interferon-induced p[ostentry inhibitor of herpesviruses and HIV-1. J Virol 92:e00709-18. doi: 10.1128/JVI.00709-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Byeon I-JL, Meng X, Jung J, Zhao G, Yang R, Ahn J, Shi J, Concel J, Aiken C, Zhang P, Gronenborn AM. 2009. Structural convergence between cryo-EM and NMR reveals intersubunit interactions critical for HIV-1 capsid function. Cell 139:780–790. doi: 10.1016/j.cell.2009.10.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. 2001. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 411:494–498. doi: 10.1038/35078107 [DOI] [PubMed] [Google Scholar]
- 145. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F. 2013. Multiplex genome engineering using CRISPR/Cas systems. Science 339:819–823. doi: 10.1126/science.1231143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Ambrose Z, Aiken C. 2014. HIV-1 uncoating: connection to nuclear entry and regulation by host proteins. Virology 454–455:371–379. doi: 10.1016/j.virol.2014.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Ingram Z, Fischer DK, Ambrose Z. 2021. Disassembling the nature of capsid: biochemical, genetic, and imaging approaches to assess HIV-1 capsid functions. Viruses 13:2237. doi: 10.3390/v13112237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Müller TG, Zila V, Müller B, Kräusslich HG. 2022. Nuclear capsid uncoating and reverse transcription of HIV-1. Annu Rev Virol 9:261–284. doi: 10.1146/annurev-virology-020922-110929 [DOI] [PubMed] [Google Scholar]
- 149. Fassati A, Goff SP. 1999. Characterization of intracellular reverse transcription complexes of moloney murine leukemia virus. J Virol 73:8919–8925. doi: 10.1128/JVI.73.11.8919-8925.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Fassati A, Goff SP. 2001. Characterization of intracellular reverse transcription complexes of human immunodeficiency virus type 1. J Virol 75:3626–3635. doi: 10.1128/JVI.75.8.3626-3635.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Brown PO, Bowerman B, Varmus HE, Bishop JM. 1987. Correct integration of retroviral DNA in vitro. Cell 49:347–356. doi: 10.1016/0092-8674(87)90287-x [DOI] [PubMed] [Google Scholar]
- 152. Bowerman B, Brown PO, Bishop JM, Varmus HE. 1989. A nucleoprotein complex mediates the integration of retroviral DNA. Genes Dev 3:469–478. doi: 10.1101/gad.3.4.469 [DOI] [PubMed] [Google Scholar]
- 153. Farnet CM, Haseltine WA. 1990. Integration of human immunodeficiency virus type 1 DNA in vitro. Proc Natl Acad Sci U S A 87:4164–4168. doi: 10.1073/pnas.87.11.4164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Farnet CM, Haseltine WA. 1991. Determination of viral proteins present in the human immunodeficiency virus type 1 preintegration complex. J Virol 65:1910–1915. doi: 10.1128/JVI.65.4.1910-1915.1991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Miller MD, Farnet CM, Bushman FD. 1997. Human immunodeficiency virus type 1 preintegration complexes: studies of organization and composition. J Virol 71:5382–5390. doi: 10.1128/JVI.71.7.5382-5390.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Balasubramaniam M, Zhou J, Addai A, Martinez P, Pandhare J, Aiken C, Dash C. 2019. PF74 inhibits HIV-1 integration by altering the composition of the preintegration complex. J Virol 93:e01741-18. doi: 10.1128/JVI.01741-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Yang Y, Luban J, Diaz-Griffero F. 2014. The fate of HIV-1 capsid: a biochemical assay for HIV-1 uncoating. Methods Mol Biol 1087:29–36. doi: 10.1007/978-1-62703-670-2_3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Márquez CL, Lau D, Walsh J, Shah V, McGuinness C, Wong A, Aggarwal A, Parker MW, Jacques DA, Turville S, Böcking T. 2018. Kinetics of HIV-1 capsid uncoating revealed by single-molecule analysis. Elife 7:e34772. doi: 10.7554/eLife.34772 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Zhang MJ, Stear JH, Jacques DA, Böcking T. 2022. Insights into HIV uncoating from single-particle imaging techniques. Biophys Rev 14:23–32. doi: 10.1007/s12551-021-00922-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. McDonald D, Vodicka MA, Lucero G, Svitkina TM, Borisy GG, Emerman M, Hope TJ. 2002. Visualization of the intracellular behavior of HIV in living cells. J Cell Biol 159:441–452. doi: 10.1083/jcb.200203150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Xu H, Franks T, Gibson G, Huber K, Rahm N, De Castillia CS, Luban J, Aiken C, Watkins S, Sluis-Cremer N, Ambrose Z. 2013. Evidence for biphasic uncoating during HIV-1 infection from a novel imaging assay. Retrovirology 10:70. doi: 10.1186/1742-4690-10-70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Mamede JI, Cianci GC, Anderson MR, Hope TJ. 2017. Early cytoplasmic uncoating is associated with infectivity of HIV-1. Proc Natl Acad Sci U S A 114:E7169–E7178. doi: 10.1073/pnas.1706245114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Li C, Burdick RC, Nagashima K, Hu WS, Pathak VK. 2021. HIV-1 cores retain their integrity until minutes before uncoating in the nucleus. Proc Natl Acad Sci U S A 118:e2019467118. doi: 10.1073/pnas.2019467118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Hübner W, Chen P, Portillo AD, Liu Y, Gordon RE, Chen BK. 2007. Sequence of human immunodeficiency virus type 1 (HIV-1) Gag localization and oligomerization monitored with live confocal imaging of a replication-competent, fluorescently tagged HIV-1. J Virol 81:12596–12607. doi: 10.1128/JVI.01088-07 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Burdick RC, Li C, Munshi M, Rawson JMO, Nagashima K, Hu W-S, Pathak VK. 2020. HIV-1 uncoats in the nucleus near sites of integration. Proc Natl Acad Sci U S A 117:5486–5493. doi: 10.1073/pnas.1920631117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Francis AC, Cereseto A, Singh PK, Shi J, Poeschla E, Engelman AN, Aiken C, Melikyan GB. 2022. Localization and functions of native and eGFP-tagged capsid proteins in HIV-1 particles. PLoS Pathog 18:e1010754. doi: 10.1371/journal.ppat.1010754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Francis AC, Marin M, Shi J, Aiken C, Melikyan GB. 2016. Time-resolved imaging of single HIV-1 uncoating in vitro and in living cells. PLoS Pathog 12:e1005709. doi: 10.1371/journal.ppat.1005709 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Francis AC, Melikyan GB. 2018. Single HIV-1 imaging reveals progression of infection through CA-dependent steps of docking at the nuclear pore, uncoating, and nuclear transport. Cell Host Microbe 23:536–548. doi: 10.1016/j.chom.2018.03.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Schifferdecker S, Zila V, Müller TG, Sakin V, Anders-Össwein M, Laketa V, Kräusslich H-G, Müller B. 2022. Direct capsid labeling of infectious HIV-1 by genetic code expansion allows detection of largely complete nuclear capsids and suggests nuclear entry of HIV-1 complexes via common routes. mBio 13:e0195922. doi: 10.1128/mbio.01959-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Platt EJ, Kozak SL, Durnin JP, Hope TJ, Kabat D. 2010. Rapid dissociation of HIV-1 from cultured cells severely limits infectivity assays, causes the inactivation ascribed to entry inhibitors, and masks the inherently high level of infectivity of Virions. J Virol 84:3106–3110. doi: 10.1128/JVI.01958-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Campbell EM, Hope TJ. 2015. HIV-1 capsid: the multifaceted key player in HIV-1 infection. Nat Rev Microbiol 13:471–483. doi: 10.1038/nrmicro3503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. von Appen A, Kosinski J, Sparks L, Ori A, DiGuilio AL, Vollmer B, Mackmull M-T, Banterle N, Parca L, Kastritis P, Buczak K, Mosalaganti S, Hagen W, Andres-Pons A, Lemke EA, Bork P, Antonin W, Glavy JS, Bui KH, Beck M. 2015. In situ structural analysis of the human nuclear pore complex. Nature 526:140–143. doi: 10.1038/nature15381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Zila V, Margiotta E, Turoňová B, Müller TG, Zimmerli CE, Mattei S, Allegretti M, Börner K, Rada J, Müller B, Lusic M, Kräusslich H-G, Beck M. 2021. Cone-shaped HIV-1 capsids are transported through intact nuclear pores. Cell 184:1032–1046. doi: 10.1016/j.cell.2021.01.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Blanco-Rodriguez G, Gazi A, Monel B, Frabetti S, Scoca V, Mueller F, Schwartz O, Krijnse-Locker J, Charneau P, Di Nunzio F, Kirchhoff F. 2020. Remodeling of the core leads HIV-1 preintegration complex into the nucleus of human lymphocytes. J Virol 94:e00135-20. doi: 10.1128/JVI.00135-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Guedán A, Donaldson CD, Caroe ER, Cosnefroy O, Taylor IA, Bishop KN, Swanstrom R. 2021. HIV-1 requires capsid remodelling at the nuclear pore for nuclear entry and integration. PLoS Pathog 17:e1009484. doi: 10.1371/journal.ppat.1009484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Dharan A, Bachmann N, Talley S, Zwikelmaier V, Campbell EM. 2020. Nuclear pore blockade reveals that HIV-1 completes reverse transcription and uncoating in the nucleus. Nat Microbiol 5:1088–1095. doi: 10.1038/s41564-020-0735-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Selyutina A, Persaud M, Lee K, KewalRamani V, Diaz-Griffero F. 2020. Nuclear import of the HIV-1 core precedes reverse transcription and uncoating. Cell Rep 32:108201. doi: 10.1016/j.celrep.2020.108201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Hulme AE, Perez O, Hope TJ. 2011. Complementary assays reveal a relationship between HIV-1 uncoating and reverse transcription. Proc Natl Acad Sci U S A 108:9975–9980. doi: 10.1073/pnas.1014522108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Cosnefroy O, Murray PJ, Bishop KN. 2016. HIV-1 capsid uncoating initiates after the first strand transfer of reverse transcription. Retrovirology 13:58. doi: 10.1186/s12977-016-0292-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Rankovic S, Varadarajan J, Ramalho R, Aiken C, Rousso I. 2017. Reverse transcription mechanically initiates HIV-1 capsid disassembly. J Virol 91:e00289-17. doi: 10.1128/JVI.00289-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Müller TG, Zila V, Peters K, Schifferdecker S, Stanic M, Lucic B, Laketa V, Lusic M, Müller B, Kräusslich H-G. 2021. HIV-1 uncoating by release of viral cDNA from capsid-like structures in the nucleus of infected cells. Elife 10:e64776. doi: 10.7554/eLife.64776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Rankovic S, Deshpande A, Harel S, Aiken C, Rousso I. 2021. HIV-1 uncoating occurs via a series of rapid biomechanical changes in the core related to individual stages of reverse transcription. J Virol 95:e00166-21. doi: 10.1128/JVI.00166-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Ma Y, Mao G, Wu G, Zhang XE. 2022. Single-particle tracking reveals the interplay between HIV-1 reverse transcription and uncoating. Anal Chem 94:2648–2654. doi: 10.1021/acs.analchem.1c05199 [DOI] [PubMed] [Google Scholar]
- 184. Luby-Phelps K. 2000. Cytoarchitecture and physical properties of cytoplasm: volume, viscosity, diffusion, intracellular surface area. Int Rev Cytol 192:189–221. doi: 10.1016/s0074-7696(08)60527-6 [DOI] [PubMed] [Google Scholar]
- 185. Fletcher DA, Mullins RD. 2010. Cell mechanics and the cytoskeleton. Nature 463:485–492. doi: 10.1038/nature08908 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Vineethakumari C, Lüders J. 2022. Microtubule anchoring: attaching dynamic polymers to cellular structures. Front Cell Dev Biol 10:867870. doi: 10.3389/fcell.2022.867870 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Sweeney HL, Holzbaur ELF. 2018. Motor proteins. Cold Spring Harb Perspect Biol 10:a021931. doi: 10.1101/cshperspect.a021931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Song Y, Brady ST. 2015. Post-translational modifications of tubulin: pathways to functional diversity of microtubules. Trends Cell Biol 25:125–136. doi: 10.1016/j.tcb.2014.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Sabo Y, Walsh D, Barry DS, Tinaztepe S, de Los Santos K, Goff SP, Gundersen GG, Naghavi MH. 2013. HIV-1 induces the formation of stable microtubules to enhance early infection. Cell Host Microbe 14:535–546. doi: 10.1016/j.chom.2013.10.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Wills JW, Craven RC. 1991. Form, function, and use of retroviral Gag proteins. AIDS 5:639–654. doi: 10.1097/00002030-199106000-00002 [DOI] [PubMed] [Google Scholar]
- 191. Reck-Peterson SL, Redwine WB, Vale RD, Carter AP. 2018. The cytoplasmic dynein transport machinery and its many cargoes. Nat Rev Mol Cell Biol 19:382–398. doi: 10.1038/s41580-018-0004-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. McKenney RJ, Huynh W, Tanenbaum ME, Bhabha G, Vale RD. 2014. Activation of cytoplasmic dynein motility by dynactin-cargo adapter complexes. Science 345:337–341. doi: 10.1126/science.1254198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Schlager MA, Hoang HT, Urnavicius L, Bullock SL, Carter AP. 2014. In vitro reconstitution of a highly processive recombinant human dynein complex. EMBO J 33:1855–1868. doi: 10.15252/embj.201488792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Hirokawa N, Noda Y, Tanaka Y, Niwa S. 2009. Kinesin superfamily motor proteins and intracellular transport. Nat Rev Mol Cell Biol 10:682–696. doi: 10.1038/nrm2774 [DOI] [PubMed] [Google Scholar]
- 195. Tang Y, Winkler U, Freed EO, Torrey TA, Kim W, Li H, Goff SP, Morse HC. 1999. Cellular motor protein KIF-4 associates with retroviral Gag. J Virol 73:10508–10513. doi: 10.1128/JVI.73.12.10508-10513.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Martinez NW, Xue X, Berro RG, Kreitzer G, Resh MD. 2008. Kinesin KIF4 regulates intracellular trafficking and stability of the human immunodeficiency virus type 1 Gag polyprotein. J Virol 82:9937–9950. doi: 10.1128/JVI.00819-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Gaudin R, de Alencar BC, Jouve M, Bèrre S, Le Bouder E, Schindler M, Varthaman A, Gobert F-X, Benaroch P. 2012. Critical role for the kinesin KIF3A in the HIV life cycle in primary human macrophages. J Cell Biol 199:467–479. doi: 10.1083/jcb.201201144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Lukic Z, Dharan A, Fricke T, Diaz-Griffero F, Campbell EM. 2014. HIV-1 uncoating is facilitated by dynein and kinesin 1. J Virol 88:13613–13625. doi: 10.1128/JVI.02219-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Dharan A, Talley S, Tripathi A, Mamede JI, Majetschak M, Hope TJ, Campbell EM, Cullen BR. 2016. KIF5B and Nup358 cooperatively mediate the nuclear import of HIV-1 during infection. PLoS Pathog 12:e1005700. doi: 10.1371/journal.ppat.1005700 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Zhang R, Mehla R, Chauhan A, Vij N. 2010. Perturbation of host nuclear membrane component RanBP2 impairs the nuclear import of human immunodeficiency virus-1 preintegration complex (DNA). PLoS ONE 5:e15620. doi: 10.1371/journal.pone.0015620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Chin CR, Perreira JM, Savidis G, Portmann JM, Aker AM, Feeley EM, Smith MC, Brass AL. 2015. Direct visualization of HIV-1 replication intermediates shows that capsid and CPSF6 modulate HIV-1 intra-nuclear invasion and integration. Cell Rep 13:1717–1731. doi: 10.1016/j.celrep.2015.10.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Achuthan V, Perreira JM, Sowd GA, Puray-Chavez M, McDougall WM, Paulucci-Holthauzen A, Wu X, Fadel HJ, Poeschla EM, Multani AS, Hughes SH, Sarafianos SG, Brass AL, Engelman AN. 2018. Capsid-CPSF6 interaction licenses nuclear HIV-1 trafficking to sites of viral DNA integration. Cell Host Microbe 24:392–404. doi: 10.1016/j.chom.2018.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Bejarano DA, Peng K, Laketa V, Börner K, Jost KL, Lucic B, Glass B, Lusic M, Müller B, Kräusslich H-G. 2019. HIV-1 nuclear import in macrophages is regulated by CPSF6-capsid interactions at the nuclear pore complex. Elife 8:e41800. doi: 10.7554/eLife.41800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Jensen D, Schekman R. 2011. COPII-mediated vesicle formation at a glance. J Cell Sci 124:1–4. doi: 10.1242/jcs.069773 [DOI] [PubMed] [Google Scholar]
- 205. Enninga J, Levay A, Fontoura BMA. 2003. Sec13 shuttles between the nucleus and the cytoplasm and stably interacts with Nup96 at the nuclear pore complex. Mol Cell Biol 23:7271–7284. doi: 10.1128/MCB.23.20.7271-7284.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Lin DH, Hoelz A. 2019. The structure of the nuclear pore complex (an update). Annu Rev Biochem 88:725–783. doi: 10.1146/annurev-biochem-062917-011901 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Lu KP, Zhou XZ. 2007. The prolyl isomerase PIN1: a pivotal new twist in phosphorylation signalling and disease. Nat Rev Mol Cell Biol 8:904–916. doi: 10.1038/nrm2261 [DOI] [PubMed] [Google Scholar]
- 208. Henning MS, Morham SG, Goff SP, Naghavi MH. 2010. PDZD8 is a novel Gag-interacting factor that promotes retroviral infection. J Virol 84:8990–8995. doi: 10.1128/JVI.00843-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Zhang S, Sodroski J. 2015. Efficient human immunodeficiency virus (HIV-1) infection of cells lacking PDZD8. Virology 481:73–78. doi: 10.1016/j.virol.2015.01.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Timney BL, Raveh B, Mironska R, Trivedi JM, Kim SJ, Russel D, Wente SR, Sali A, Rout MP. 2016. Simple rules for passive diffusion through the nuclear pore complex. J Cell Biol 215:57–76. doi: 10.1083/jcb.201601004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Kalita J, Kapinos LE, Lim RYH. 2021. On the asymmetric partitioning of nucleocytoplasmic transport - recent insights and open questions. J Cell Sci 134:jcs240382. doi: 10.1242/jcs.240382 [DOI] [PubMed] [Google Scholar]
- 212. Zhong H, Takeda A, Nazari R, Shio H, Blobel G, Yaseen NR. 2005. Carrier-independent nuclear import of the transcription factor PU.1 via RanGTP-stimulated binding to Nup153. J Biol Chem 280:10675–10682. doi: 10.1074/jbc.M412878200 [DOI] [PubMed] [Google Scholar]
- 213. Meehan AM, Saenz DT, Guevera R, Morrison JH, Peretz M, Fadel HJ, Hamada M, van Deursen J, Poeschla EM. 2014. A cyclophilin homology domain-independent role for Nup358 in HIV-1 infection. PLoS Pathog 10:e1003969. doi: 10.1371/journal.ppat.1003969 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Matreyek KA, Engelman A. 2011. The requirement for nucleoporin NUP153 during human immunodeficiency virus type 1 infection is determined by the viral capsid. J Virol 85:7818–7827. doi: 10.1128/JVI.00325-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Dicks MDJ, Betancor G, Jimenez-Guardeño JM, Pessel-Vivares L, Apolonia L, Goujon C, Malim MH. 2018. Multiple components of the nuclear pore complex interact with the amino-terminus of MX2 to facilitate HIV-1 restriction. PLoS Pathog 14:e1007408. doi: 10.1371/journal.ppat.1007408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Christ F, Thys W, De Rijck J, Gijsbers R, Albanese A, Arosio D, Emiliani S, Rain J-C, Benarous R, Cereseto A, Debyser Z. 2008. Transportin-SR2 imports HIV into the nucleus. Curr Biol 18:1192–1202. doi: 10.1016/j.cub.2008.07.079 [DOI] [PubMed] [Google Scholar]
- 217. Krishnan L, Matreyek KA, Oztop I, Lee K, Tipper CH, Li X, Dar MJ, Kewalramani VN, Engelman A. 2010. The requirement for cellular transportin 3 (TNPO3 or TRN-SR2) during infection maps to human immunodeficiency virus type 1 capsid and not Integrase. J Virol 84:397–406. doi: 10.1128/JVI.01899-09 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. De Iaco A, Santoni F, Vannier A, Guipponi M, Antonarakis S, Luban J. 2013. TNPO3 protects HIV-1 replication from CPSF6-mediated capsid stabilization in the host cell cytoplasm. Retrovirology 10:20. doi: 10.1186/1742-4690-10-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Fricke T, Valle-Casuso JC, White TE, Brandariz-Nuñez A, Bosche WJ, Reszka N, Gorelick R, Diaz-Griffero F. 2013. The ability of TNPO3-depleted cells to inhibit HIV-1 infection requires CPSF6. Retrovirology 10:46. doi: 10.1186/1742-4690-10-46 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Rüegsegger U, Beyer K, Keller W. 1996. Purification and characterization of human cleavage factor Im involved in the 3' end processing of messenger RNA precursors. J Biol Chem 271:6107–6113. doi: 10.1074/jbc.271.11.6107 [DOI] [PubMed] [Google Scholar]
- 221. Maertens GN, Cook NJ, Wang W, Hare S, Gupta SS, Öztop I, Lee K, Pye VE, Cosnefroy O, Snijders AP, KewalRamani VN, Fassati A, Engelman A, Cherepanov P. 2014. Structural basis for nuclear import of splicing factors by human transportin 3. Proc Natl Acad Sci U S A 111:2728–2733. doi: 10.1073/pnas.1320755111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Jang S, Cook NJ, Pye VE, Bedwell GJ, Dudek AM, Singh PK, Cherepanov P, Engelman AN. 2019. Differential role for phosphorylation in alternative polyadenylation function versus nuclear import of SR-like protein CPSF6. Nucleic Acids Res 47:4663–4683. doi: 10.1093/nar/gkz206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Sowd GA, Serrao E, Wang H, Wang W, Fadel HJ, Poeschla EM, Engelman AN. 2016. A critical role for alternative polyadenylation factor CPSF6 in targeting HIV-1 integration to transcriptionally active chromatin. Proc Natl Acad Sci U S A 113:E1054–E1063. doi: 10.1073/pnas.1524213113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. De Houwer S, Demeulemeester J, Thys W, Rocha S, Dirix L, Gijsbers R, Christ F, Debyser Z. 2014. The HIV-1 integrase mutant R263A/K264A is 2-fold defective for TRN-SR2 binding and viral nuclear import. J Biol Chem 289:25351–25361. doi: 10.1074/jbc.M113.533281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Janssens J, Blokken J, Lampi Y, De Wit F, Zurnic Bonisch I, Nombela I, Van de Velde P, Van Remoortel B, Gijsbers R, Christ F, Debyser Z. 2021. CRISPR/Cas9-induced mutagenesis corroborates the role of transportin-SR2 in HIV-1 nuclear import. Microbiol Spectr 9:e0133621. doi: 10.1128/Spectrum.01336-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Henning MS, Dubose BN, Burse MJ, Aiken C, Yamashita M. 2014. In vivo functions of CPSF6 for HIV-1 as revealed by HIV-1 capsid evolution in HLA-B27-positive subjects. PLoS Pathog 10:e1003868. doi: 10.1371/journal.ppat.1003868 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Albanese M, Ruhle A, Mittermaier J, Mejías-Pérez E, Gapp M, Linder A, Schmacke NA, Hofmann K, Hennrich AA, Levy DN, Humpe A, Conzelmann KK, Hornung V, Fackler OT, Keppler OT. 2022. Rapid, efficient and activation-neutral gene editing of polyclonal primary human resting CD4+ T cells allows complex functional analyses. Nat Methods 19:81–89. doi: 10.1038/s41592-021-01328-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228. Labokha AA, Gradmann S, Frey S, Hülsmann BB, Urlaub H, Baldus M, Görlich D. 2013. Systematic analysis of barrier-forming FG hydrogels from Xenopus nuclear pore complexes. EMBO J 32:204–218. doi: 10.1038/emboj.2012.302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Chug H, Trakhanov S, Hülsmann BB, Pleiner T, Görlich D. 2015. Crystal structure of the metazoan Nup62•Nup58•Nup54 nucleoporin complex. Science 350:106–110. doi: 10.1126/science.aac7420 [DOI] [PubMed] [Google Scholar]
- 230. Monette A, Panté N, Mouland AJ. 2011. HIV-1 remodels the nuclear pore complex. J Cell Biol 193:619–631. doi: 10.1083/jcb.201008064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Bhargava A, Williart A, Maurin M, Davidson PM, Jouve M, Piel M, Lahaye X, Manel N. 2021. Inhibition of HIV infection by structural proteins of the inner nuclear membrane is associated with reduced chromatin dynamics. Cell Rep 36:109763. doi: 10.1016/j.celrep.2021.109763 [DOI] [PubMed] [Google Scholar]
- 232. Engelman AN, Maertens GN. 2018. Virus-host interactions in retrovirus integration, p 163–198. In Parent LJ (ed), Retrovirus-cell interactions. Academic Press, San Diego, CA. [Google Scholar]
- 233. Bedwell GJ, Engelman AN. 2021. Factors that mold the nuclear landscape of HIV-1 integration. Nucleic Acids Res 49:621–635. doi: 10.1093/nar/gkaa1207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Schröder ARW, Shinn P, Chen H, Berry C, Ecker JR, Bushman F. 2002. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110:521–529. doi: 10.1016/S0092-8674(02)00864-4 [DOI] [PubMed] [Google Scholar]
- 235. Wang GP, Ciuffi A, Leipzig J, Berry CC, Bushman FD. 2007. HIV integration site selection: analysis by massively parallel pyrosequencing reveals association with epigenetic modifications. Genome Res 17:1186–1194. doi: 10.1101/gr.6286907 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Francis AC, Marin M, Singh PK, Achuthan V, Prellberg MJ, Palermino-Rowland K, Lan S, Tedbury PR, Sarafianos SG, Engelman AN, Melikyan GB. 2020. HIV-1 replication complexes accumulate in nuclear speckles and integrate into speckle-associated genomic domains. Nat Commun 11:3505. doi: 10.1038/s41467-020-17256-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Rheinberger M, Costa AL, Kampmann M, Glavas D, Shytaj IL, Sreeram S, Penzo C, Tibroni N, Garcia-Mesa Y, Leskov K, Fackler OT, Vlahovicek K, Karn J, Lucic B, Herrmann C, Lusic M. 2023. Genomic profiling of HIV-1 integration in microglia cells links viral integration to the topologically associated domains. Cell Rep 42:112110. doi: 10.1016/j.celrep.2023.112110 [DOI] [PubMed] [Google Scholar]
- 238. Marini B, Kertesz-Farkas A, Ali H, Lucic B, Lisek K, Manganaro L, Pongor S, Luzzati R, Recchia A, Mavilio F, Giacca M, Lusic M. 2015. Nuclear architecture dictates HIV-1 integration site selection. Nature 521:227–231. doi: 10.1038/nature14226 [DOI] [PubMed] [Google Scholar]
- 239. van Steensel B, Belmont AS. 2017. Lamina-associated domains: links with chromosome architecture, heterochromatin, and gene repression. Cell 169:780–791. doi: 10.1016/j.cell.2017.04.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Scoca V, Morin R, Collard M, Tinevez J-Y, Di Nunzio F. 2023. HIV-induced membraneless organelles orchestrate post-nuclear entry steps. J Mol Cell Biol 14:mjac060. doi: 10.1093/jmcb/mjac060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Cherepanov P, Maertens G, Proost P, Devreese B, Van Beeumen J, Engelborghs Y, De Clercq E, Debyser Z. 2003. HIV-1 integrase forms stable tetramers and associates with LEDGF/p75 protein in human cells. J Biol Chem 278:372–381. doi: 10.1074/jbc.M209278200 [DOI] [PubMed] [Google Scholar]
- 242. Ciuffi A, Llano M, Poeschla E, Hoffmann C, Leipzig J, Shinn P, Ecker JR, Bushman F. 2005. A role for LEDGF/p75 in targeting HIV DNA integration. Nat Med 11:1287–1289. doi: 10.1038/nm1329 [DOI] [PubMed] [Google Scholar]
- 243. Shun MC, Raghavendra NK, Vandegraaff N, Daigle JE, Hughes S, Kellam P, Cherepanov P, Engelman A. 2007. LEDGF/p75 functions downstream from preintegration complex formation to effect gene-specific HIV-1 integration. Genes Dev 21:1767–1778. doi: 10.1101/gad.1565107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Wu X, Li Y, Crise B, Burgess SM. 2003. Transcription start regions in the human genome are favored targets for MLV integration. Science 300:1749–1751. doi: 10.1126/science.1083413 [DOI] [PubMed] [Google Scholar]
- 245. Singh PK, Plumb MR, Ferris AL, Iben JR, Wu X, Fadel HJ, Luke BT, Esnault C, Poeschla EM, Hughes SH, Kvaratskhelia M, Levin HL. 2015. LEDGF/p75 interacts with mRNA splicing factors and targets HIV-1 integration to highly spliced genes. Genes Dev 29:2287–2297. doi: 10.1101/gad.267609.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Ocwieja KE, Brady TL, Ronen K, Huegel A, Roth SL, Schaller T, James LC, Towers GJ, Young JAT, Chanda SK, König R, Malani N, Berry CC, Bushman FD. 2011. HIV integration targeting: a pathway involving transportin-3 and the nuclear pore protein RanBP2. PLoS Pathog 7:e1001313. doi: 10.1371/journal.ppat.1001313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Koh Y, Wu X, Ferris AL, Matreyek KA, Smith SJ, Lee K, KewalRamani VN, Hughes SH, Engelman A. 2013. Differential effects of human immunodeficiency virus type 1 capsid and cellular factors nucleoporin 153 and LEDGF/p75 on the efficiency and specificity of viral DNA integration. J Virol 87:648–658. doi: 10.1128/JVI.01148-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Llano M, Vanegas M, Fregoso O, Saenz D, Chung S, Peretz M, Poeschla EM. 2004. LEDGF/p75 determines cellular trafficking of diverse lentiviral but not murine oncoretroviral integrase proteins and is a component of functional lentiviral preintegration complexes. J Virol 78:9524–9537. doi: 10.1128/JVI.78.17.9524-9537.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Busschots K, Vercammen J, Emiliani S, Benarous R, Engelborghs Y, Christ F, Debyser Z. 2005. The interaction of LEDGF/p75 with integrase is lentivirus-specific and promotes DNA binding. J Biol Chem 280:17841–17847. doi: 10.1074/jbc.M411681200 [DOI] [PubMed] [Google Scholar]
- 250. Cherepanov P. 2007. LEDGF/p75 interacts with divergent lentiviral Integrases and modulates their enzymatic activity in vitro. Nucleic Acids Res 35:113–124. doi: 10.1093/nar/gkl885 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Li W, Singh PK, Sowd GA, Bedwell GJ, Jang S, Achuthan V, Oleru AV, Wong D, Fadel HJ, Lee K, KewalRamani VN, Poeschla EM, Herschhorn A, Engelman AN, Goff SP. 2020. CPSF6-dependent targeting of speckle-associated domains distinguishes primate from nonprimate lentiviral integration. mBio 11:e02254-20. doi: 10.1128/mBio.02254-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Singh PK, Bedwell GJ, Engelman AN. 2022. Spatial and genomic correlates of HIV-1 integration site targeting. Cells 11:655. doi: 10.3390/cells11040655 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Rensen E, Mueller F, Scoca V, Parmar JJ, Souque P, Zimmer C, Di Nunzio F. 2021. Clustering and reverse transcription of HIV-1 genomes in nuclear niches of macrophages. EMBO J 40:e105247. doi: 10.15252/embj.2020105247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Banani SF, Lee HO, Hyman AA, Rosen MK. 2017. Biomolecular condensates: organizers of cellular biochemistry. Nat Rev Mol Cell Biol 18:285–298. doi: 10.1038/nrm.2017.7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Shin Y, Brangwynne CP. 2017. Liquid phase condensation in cell physiology and disease. Science 357:eaaf4382. doi: 10.1126/science.aaf4382 [DOI] [PubMed] [Google Scholar]
- 256. Strom AR, Brangwynne CP. 2019. The liquid nucleome - phase transitions in the nucleus at a glance. J Cell Sci 132:jcs235093. doi: 10.1242/jcs.235093 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Ay S, Di Nunzio F. 2023. HIV-induced CPSF6 condensates. J Mol Biol:168094. doi: 10.1016/j.jmb.2023.168094 [DOI] [PubMed] [Google Scholar]
- 258. Jalihal AP, Pitchiaya S, Xiao L, Bawa P, Jiang X, Bedi K, Parolia A, Cieslik M, Ljungman M, Chinnaiyan AM, Walter NG. 2020. Multivalent proteins rapidly and reversibly phase-separate upon osmotic cell volume change. Mol Cell 79:978–990. doi: 10.1016/j.molcel.2020.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Kawachi T, Masuda A, Yamashita Y, Takeda J-I, Ohkawara B, Ito M, Ohno K. 2021. Regulated splicing of large Exons is linked to phase-separation of vertebrate transcription factors. EMBO J 40:e107485. doi: 10.15252/embj.2020107485 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Hultquist JF, Hiatt J, Schumann K, McGregor MJ, Roth TL, Haas P, Doudna JA, Marson A, Krogan NJ. 2019. CRISPR-Cas9 genome engineering of primary CD4+ T cells for the interrogation of HIV-host factor interactions. Nat Protoc 14:1–27. doi: 10.1038/s41596-018-0069-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Hiatt J, Cavero DA, McGregor MJ, Zheng W, Budzik JM, Roth TL, Haas KM, Wu D, Rathore U, Meyer-Franke A, Bouzidi MS, Shifrut E, Lee Y, Kumar VE, Dang EV, Gordon DE, Wojcechowskyj JA, Hultquist JF, Fontaine KA, Pillai SK, Cox JS, Ernst JD, Krogan NJ, Marson A. 2021. Efficient generation of isogenic primary human myeloid cells using CRISPR-Cas9 ribonucleoproteins. Cell Rep 35:109105. doi: 10.1016/j.celrep.2021.109105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Bester SM, Wei G, Zhao H, Adu-Ampratwum D, Iqbal N, Courouble VV, Francis AC, Annamalai AS, Singh PK, Shkriabai N, Van Blerkom P, Morrison J, Poeschla EM, Engelman AN, Melikyan GB, Griffin PR, Fuchs JR, Asturias FJ, Kvaratskhelia M. 2020. Structural and mechanistic bases for a potent HIV-1 capsid inhibitor. Science 370:360–364. doi: 10.1126/science.abb4808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Link JO, Rhee MS, Tse WC, Zheng J, Somoza JR, Rowe W, Begley R, Chiu A, Mulato A, Hansen D, Singer E, Tsai LK, Bam RA, Chou C-H, Canales E, Brizgys G, Zhang JR, Li J, Graupe M, Morganelli P, Liu Q, Wu Q, Halcomb RL, Saito RD, Schroeder SD, Lazerwith SE, Bondy S, Jin D, Hung M, Novikov N, Liu X, Villaseñor AG, Cannizzaro CE, Hu EY, Anderson RL, Appleby TC, Lu B, Mwangi J, Liclican A, Niedziela-Majka A, Papalia GA, Wong MH, Leavitt SA, Xu Y, Koditek D, Stepan GJ, Yu H, Pagratis N, Clancy S, Ahmadyar S, Cai TZ, Sellers S, Wolckenhauer SA, Ling J, Callebaut C, Margot N, Ram RR, Liu Y-P, Hyland R, Sinclair GI, Ruane PJ, Crofoot GE, McDonald CK, Brainard DM, Lad L, Swaminathan S, Sundquist WI, Sakowicz R, Chester AE, Lee WE, Daar ES, Yant SR, Cihlar T. 2020. Clinical targeting of HIV capsid protein with a long-acting small molecule. Nature 584:614–618. doi: 10.1038/s41586-020-2443-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Cherepanov P, Ambrosio ALB, Rahman S, Ellenberger T, Engelman A. 2005. Structural basis for the recognition between HIV-1 integrase and transcriptional coactivator p75. Proc Natl Acad Sci U S A 102:17308–17313. doi: 10.1073/pnas.0506924102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Christ F, Voet A, Marchand A, Nicolet S, Desimmie BA, Marchand D, Bardiot D, Van der Veken NJ, Van Remoortel B, Strelkov SV, De Maeyer M, Chaltin P, Debyser Z. 2010. Rational design of small-molecule inhibitors of the LEDGF/p75-integrase interaction and HIV replication. Nat Chem Biol 6:442–448. doi: 10.1038/nchembio.370 [DOI] [PubMed] [Google Scholar]
- 266. Gupta K, Turkki V, Sherrill-Mix S, Hwang Y, Eilers G, Taylor L, McDanal C, Wang P, Temelkoff D, Nolte RT, Velthuisen E, Jeffrey J, Van Duyne GD, Bushman FD. 2016. Structural basis for inhibitor-induced aggregation of HIV integrase. PLoS Biol 14:e1002584. doi: 10.1371/journal.pbio.1002584 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Gupta K, Allen A, Giraldo C, Eilers G, Sharp R, Hwang Y, Murali H, Cruz K, Janmey P, Bushman F, Van Duyne GD. 2021. Allosteric HIV integrase inhibitors promote formation of inactive branched polymers via homomeric carboxy-terminal domain interactions. Structure 29:213–225. doi: 10.1016/j.str.2020.12.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Jurado KA, Wang H, Slaughter A, Feng L, Kessl JJ, Koh Y, Wang W, Ballandras-Colas A, Patel PA, Fuchs JR, Kvaratskhelia M, Engelman A. 2013. Allosteric integrase inhibitor potency is determined through the inhibition of HIV-1 particle maturation. Proc Natl Acad Sci U S A 110:8690–8695. doi: 10.1073/pnas.1300703110 [DOI] [PMC free article] [PubMed] [Google Scholar]