HM Medical Clinic

 

Chem.ch.huji.ac.il

For reprint orders, please contact [email protected] Chemistry
Review 2015/04/28 Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Future Med. Chem.
Protein–protein interactions (PPI) are essential in every step of the HIV replication Koushik Chandra1,2, Michal cycle. Mapping the interactions between viral and host proteins is a fundamental Maes1 & Assaf Friedler*,11Institute of Chemistry, The Hebrew target for the design and development of new therapeutics. In this review, we focus University of Jerusalem, Safra Campus, on rational development of anti-HIV-1 peptides based on mapping viral–host and Givat Ram, Jerusalem 91904, Israel viral–viral protein interactions all across the HIV-1 replication cycle. We also discuss the 2Department of Chemistry, Midnapore mechanism of action, specificity and stability of these peptides, which are designed College, West Bengal, India to inhibit PPI. Some of these peptides are excellent tools to study the mechanisms of *Author for correspondence: [email protected] PPI in HIV-1 replication cycle and for the development of anti-HIV-1 drug leads that modulate PPI.
Protein–protein interactions in the
HHPID reports 15 essential HIV-1 pro- HIV-1 replication cycle
teins [25,31,41–44] (Figures 1 & 2). Three funda- Mapping the interactions between proteins mental proteins (Gag, Pol, Env) are encoded derived from host and pathogen origins is by the HIV-1 genome and they undergo pro-essential for understanding the molecu- teolysis to form the mature proteins. Four lar mechanisms of host–pathogen interac- structural proteins, matrix (MA), capsid tions [1–4]. Protein–protein interactions (CA), nucleocapsid (NC) and p6, are prod-(PPI) play a crucial role in the replication of ucts of the proteolysis of Gag. Env proteolysis HIV-1 [5–24]. HIV-1 infection results in an results in the envelope proteins gp120 and interplay between viral and host proteins or gp41 [45,46]. Pol encodes three enzymes: pro-homodimeric/oligomeric viral protein inter- tease (PR), reverse transcriptase (RT) and actions, resulting in a complex interaction integrase (IN). Encapsulated within the network between various proteins [25,26]. virus particle, the three Pol proteins play key The HIV-1-Human Protein Interaction functions in the viral replication upon infec- Database (HHPID) identified 1435 human tion. The remaining proteins (Vif, Vpr, Nef, genes encoding 1448 human proteins that Tat, Rev, Vpu) are accessory proteins [47–50]. interact with HIV-1 proteins, resulting in The database shows 43 different direct inter-2589 unique HIV-1-host protein interac- actions of HIV-1 proteins with human pro- tions [27–33]. Thirty two percent of these are teins based on activity, binding, inhibition, direct physical interactions as revealed from cleavage, complexation, modulation, deglyco-binding studies and 68% are indirect interac- sylation and upregulation. Only a part of these tions such as upregulation through activation interactions are targets for peptide inhibitors of signaling pathways. The database reveals and will be discussed here (Figure 2).
that numerous human proteins interact with more than one HIV-1 protein. Using a quan- Peptides as a tool to study PPI
titative scoring system termed mass spec- Understanding PPI requires thorough struc- trometric interaction statistics (MiST), 497 tural, biophysical and biochemical character-HIV-human PPIs involving 435 individual ization using recombinant proteins. However, human proteins and 18 viral proteins have a major hurdle is the expression and purifica- been identified [25,34–40].
tion of the interacting proteins. Some proteins 10.4155/FMC.15.46 2015 Future Science Ltd Future Med. Chem. (2015) 7(8), 1055–1077
Review Chandra, Maes & Friedler Figure 1. HIV-1 replication cycle with the essential viral proteins highlighted.
are insoluble or toxic to the expressing host, resulting present an overview of peptides derived from PPI from in low yields that hamper structural and quantitative different stages of the HIV-1 replication cycle [71] and studies. Using peptides for these studies provide many their implications for anti-HIV-1 drug design.
advantages relative to the recombinant proteins. Pep-tides derived from the interacting proteins enable deter- PART I: interactions between viral & host
mination of the specific interaction sites, the affinity proteins
and thermodynamic contribution (enthalpy vs entropy) Interactions between the viral capsid & host
in PPI [51–54]. Chemical synthesis of the peptides makes membrane proteins
it possible to overcome the expression and purification The initial contact between the virus and the host cell
related problems of protein production [55–57]. This is made between the viral glycoprotein gp120 (originat-
makes it technically convenient to study the PPI via ing from the Env polyprotein, PDB: 3DNL, Figure 3A)
a full-length protein and a peptide derived from the and the cell surface receptor CD4 [72,73]. CD4 is a host
complementary protein in addition to the interaction glycoprotein expressed on the surface of T helper cells,
between the two full-length proteins. Peptides derived regulatory T cells, macrophages, monocytes and den-
from binding interfaces may bind weaker than the par-
dritic cells. The binding of a highly conserved, nong- ent protein, partly due to loss of secondary structure. lycosylated region of gp120 to CD4 results in the viral Modifications such as post-translational modifications insertion into the host membrane [74–76]. Upon associa-(e.g., acetylation or phosphorylation) [58,59], labeling tion with another viral envelope protein, gp41 (PDB: (e.g., fluorescein or biotin) or incorporation of non- 2ZFC, Figure 3B), which mediates viral entry through natural amino acids can be inserted specifically into a membrane fusion, binding to CD4 occurs. This results protein sequence only using chemical peptide synthe- in a conformational change that allows gp120 to bind sis [60,61]. Peptides are an excellent model for binding to the coreceptors CCR5 or CXCR4 [77,78]; belonging studies of protein domains. Upon binding, they can to the family of G protein-coupled receptors and che-undergo conformational change mimicking the native mokine receptors [79–81]. The viral insertion causes an binding interface [62,63]. This makes peptides a useful additional conformational change in the heptad repeat tool for discovering drug leads by modulating (either regions (HR1 and HR2) of gp41 [82], resulting in the activating or inhibiting) PPI [64–70]. In this review, we entry of the viral capsid into the host cell via a fusion Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review Figure 2. Selected PPI of HIV-1 proteins. (A) An interaction map between direct interactions of HIV-1-proteins (green)
with human proteins (purple). (B) An interaction map between direct interactions of viral proteins (green). Both the
interactions are involved in protein–protein interactions that served as basis for developing inhibitory peptides.
For color images please see online www.future-science.com/doi/full/10.4155/FMC.15.46
pore [83]. This leads to the generation of epitopes for HIV-1 gp120 & CXCR4 interactionsneutralizing antibodies that prevent chemokine recep- One of the functions of gp120 is tethering of the tor binding [84,85]. Two inhibitors of fusion and entry virus to the cellular co-receptor CXCR4. CXCR4 are currently used in the clinic. Approved in 2003, the binds the bridging sheet and V3 loop of gp120 [92,93]. 36-mer peptide inhibitor T20 (Enfuvirtide) blocks a The binding between CXCR4 and gp120 involves a critical conformational change in gp41 responsible for conformational rearrangement of gp120. The soluble membrane fusion [86]. Maraviroc is a small molecule synthetic peptide, CX4-M1, functionally mimics the antiretroviral drug, approved in 2007, which inhibits HIV-1 co-receptor CXCR4 [85,94]. The interaction the interaction between gp120 and CCR5 [86].
interface between gp120 and its cellular co-receptor partner CXCR4 is between the V3 loop of gp120 Peptides derived from HIV-1 Env & host proteins and the extracellular loops (ECLs) of CXCR4. The CX4-M1 peptides are derived from the ECL region The HIV inhibiting peptide database (HIPdb) reveals of CXCR4 from different HIV1 strains and bind-110 HIV inhibitory peptides that target the interac- ing was determined via direct ELISA [95]. The bind- tions of the viral Env proteins. They aim to prevent the ing affinities between the peptides and the protein interactions between the virus and cellular cofactors were measured by surface plasmon resonance (SPR) by binding either viral envelope proteins or host pro- (Table 1C). To confirm specific binding, CX4-M1 teins [87,88]. Table 1A shows the best HIV-1 inhibitory was competed with a specific antibody, mAb447–peptide based on the prediction of antigenicity method 52D, that recognizes the V3 loop of gp120. A peptide for inhibiting Env proteins. The HIV-1 envelope pro- binding assay with CX4-M1 and V3 loop peptides tein gp41 fragment peptide (residues 568–588) is derived from the N-heptad region of gp41 Env ectodo- main [89]. It specifically binds the phospholipid mem- gp120: HIV-1 envelope glycoprotein encoded by the brane thereby inhibiting the viral-cell fusion process. HIV env gene. The virus entry into cel s is anchored by Microcalorimetric titrations revealed that a 22-resides gp120. The process is mediated by the binding of gp120, which is exposed on the surface of the HIV envelope to tyrosine-sulfated peptide (S22 peptide) derived from specific cell surface receptors such as CD4, heparan sulfate the N-terminus of CCR5 showed a strong interac- proteoglycan. The change in the conformation of gp120 tion with the gp120-CD4 complex with K = 2.2 μM triggers fusion between the viral and host cell membranes.
(Table 1B). The process is both entropically and enthal- Integrase: Viral enzyme encoded by HIV-1, which catalyzes pically favorable. No binding was observed between the integration of the viral cDNA into the host cell genome. the gp120-CD4 complex and an identical peptide IN performs two enzymatic activities: 3′-end processing in lacking the sulfated tyrosine residues the cytoplasm and strand transfer in the nucleus.
future science group Review Chandra, Maes & Friedler Figure 3. X-ray crystal structures of some HIV-1 proteins. (A) HIV-1 gp120 trimer (PDB: 3DNL) [279]; (B) NTD of HIV-1
gp41 trimer (PDB: 2ZFC) [280]; and (C) HIV-1 RT with DNA (PDB: 3V4I) [281].
confirmed that the V3 loop is the crucial part of the RT is a heterodimeric protein with two asymmetric co-receptor binding site of gp120.
chains termed p51 and p66 [97]. HIV-1 RT has three main activities: RNA-directed DNA polymerization, The viral enzyme reverse transcriptase
DNA-directed RNA polymerization and exonuclease HIV-1 reverse transcriptase (RT) produces a viral cDNA via degradation of RNA [98]. RT is the target of numer-based on the viral RNA (PDB: 3V4I, Figure 3C). The ous small molecule antiretroviral drugs used in the DNA is later integrated into the host cell genome [96]. clinic [99]. AZT is a Nucleoside analog RT Inhibitor Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review Table 1. Peptides that inhibit viral entry.
(A) Env and CD4 interaction
(B) gp120 and CCR5 interaction
MDYQVSSPIY(SO -)DINY(SO -)YTSEPSQK (C) gp120 and CXCR4 interaction (ECL1)CX4-M1
PPI: Protein–protein interaction.
(NRTI) that acts as a chain terminator of growing viral cDNA into the host genome (Figure 1) [111–115]. It DNA strand and was approved as an anti-HIV drug has three functional domains responsible for integra-in 1987. In 1996, Nevirapine was approved as the first tion process: the N-terminal domain (NTD), the cata-non-nucleoside RT inhibitor (NNRTI) that inhibits lytic core domain (CCD) and the C-terminal domain the RT polymerization activity.
(CTD) [116,117]. IN has two enzymatic activities: first, 3′-end processing in the cytoplasm [111,118] in which two The HIV-1 RT & A3G interaction IN dimers [119] bind the long terminal repeats (LTR) of During reverse transcription, the human cytidine deam- the viral DNA and remove a pGT dinucleotide from the inase APOBEC3G (A3G) eliminates HIV-1 infection 3′-end of each strand. After nuclear transport [120,121], by inducing deamination of the cytosine residues to the strand transfer reaction is carried out by an IN tet-uracil in the negative viral DNA strand [100–108]. Using ramer [122–124] resulting in integration of the viral DNA a cell-based co-immunoprecipitation (coIP) assay, the into the host genome. Finally, the single-stranded gaps direct interaction of A3G with RT was detected both in transfected cells and in the produced viruses. No other viral components are needed for this interaction.
Deletion analysis with a series of T7-tagged RT-dele-
tion mutants (T7-RT1–243, T7-RT1–323 and T7-RT1–439)
determined that the RT-binding domain is located
at the N-terminal region of A3G65–132 [101,109,110]. The
polypeptide A3G65–132 inhibited the interaction between
A3G and the viral RT (PDB: 3VOW, Figure 4A,
Table 2A) [101]. The RT-binding polypeptide inhibited
the anti-HIV effect of A3G on RT. Competitive coIP Figure 4. Peptides derived from human APOBEC3G.
in cells co-expressing both RT and A3G using sev- (A) Crystal structure of human A3G. The RT-binding
eral A3G derived polypeptides showed that A3G65–132 A3G 65–132 peptide is shown in cyan (PDB:3VOW) [282], (B) Crystal structure of APOBEC3G catalytic core
significantly disrupted the A3G-RT binding.
domain (CCD); the Vif-interactions regions are: A3G 211–225 (magenta), A3G 263–278 (cyan), A3G 331–345 Interactions of the HIV-1 integrase
(green) and A3G 353–367 (red) (PDB:3IR2) [283]. HIV-1 integrase (IN) plays one of the key roles in the viral For color images please see online www.future-science.
replication cycle by integrating the reverse transcribed com/doi/full/10.4155/FMC.15.46 future science group Review Chandra, Maes & Friedler Table 2. Peptides that inhibit the viral enzymes reverse transcriptase and integrase.
(A) RT & A3G
(B) IN dimerization IN 93–107(INH1)
IN 129–139(NL9) IN 129–139W131A IN 171–187(α5) IN 196–210(α6) IN 196–206(α6S) IN 151–175(K156 E (B) Cellular partner LEDGF/p75 354–378
LEDGF/p75 355–377 LEDGF/p75 361–370 LEDGF/p75 362–369 LEDGF/p75 402–413 (C) Phage display
†Inverted sequence with d-amino acids.
PPI: Protein–protein interaction.
between the viral DNA and target DNA are repaired by tors. Some of the peptides (IN 95–109, IN 97–108, IN 171–187 the host DNA repair machinery [125–127]. The equilib- and IN 196–210) showed very mild IC for both 3′-pro- rium between dimeric and tetrameric IN is of extreme cessing and strand transfer in vitro. IN 147–175, which is importance in the integration process, making it an derived from IN, inhibited IN at 600 μM concentration attractive target for drug design [69]. In 2007, Ralte- by partly blocking the active site. The peptide inhib- gravir was the first IN inhibitor approved for clinical ited the catalytic activity of IN by binding it through use. Another IN inhibitor, Elvitegravir, was approved a protein-peptide coiled-coil structure [130,133–135]. Two for clinical use in 2012 [128,129]. Both inhibitors block peptides derived from the α1 and α5 helices of the IN by binding directly to the IN-DNA complex formed CCD, (INH1 and INH5) specifically bound to the during the integration of the viral DNA into the host dimerization interface of the CCD of IN [136]. The cell genome [128,129].
IC for 3′- processing by INH1 was 250 μM and by INH5 was 11 μM. By inhibiting the 3′-endonuclase Peptides derived from the dimerization activity of IN with IC values in the low micromolar range, three peptides (α1, α5, α6) also inhibited the IN The dimerization interface of IN is an excellent start- dimerization [137]. The truncated peptide (NL6–5) and ing point for peptides that would inhibit dimer forma- retro-inverso-peptides (RDNL6, RDNL9) retained the tion [130–132]. Several peptides have been designed (PDB: inhibitory activity by disrupting the IN dimer and tet-3L3U, Figure 5A, Table 2B) but due to relatively low ramer formation [138,139] All the peptides were derived binding affinity to IN, they did not succeed in disrupt- from the CCD of IN, which is the only domain that ing the dimeric IN and hence were not efficient inhibi- mediates IN dimerization (Figure 5A) [138,139].
Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review Figure 5. Peptides derived from domains of HIV-1 integrase. (A) Crystal structure of HIV-1 IN catalytic core domain
(CCD, beige and green) dimer illustrates the important regions of the IN-dimerization interface from where
peptides were derived: IN 93–107 (INH1, α1, NL6) (magenta), IN 171–208 (α5, α6, α6s) (magenta) (PDB:3L3U) [284].
(B) Crystal structure of dimeric IN CCD and LEDGF/p75 IBD (gray) showing interacting regions: LEDGF/p75 354–378
(cyan), LEDGF/p75 361–370 (red), LEDGF/p75 402–411 (magenta) [152].
For color images please see online www.future-science.com/doi/full/10.4155/FMC.15.46
Peptides derived from cellular proteins that LEDGF/p75402–413 are important for optimal binding and inhibition of IN (Figure 5B) [69,153–156]. A library of Targeting host proteins is risky since it may affect cell cyclic peptides (CPs) derived from LEDGF/p75361–370 viability and produce undesired toxicity. Therefore, the was screened for in vitro IN binding and inhibition [155]. best strategy is to study the interactions between IN and One of these peptides, c(MZ4–1) was a potent and sta-host proteins by finding peptides derived from the IN- ble inhibitor of IN in vitro. NMR and docking studies binding region of the cellular proteins. These peptides revealed that c(MZ4–1) possessed a conformation almost will potentially bind the viral protein and inhibit the identical to the parent IN-binding loop from the IBD of interaction with less potential for undesired side effects.
LEDGF/p75. An AlphaScreen assay with these peptides also accounted for IN-LEDGF/p75 interaction [157].
The IN-LEDGF/p75 interaction A random peptide phage display strategy was In addition to binding the viral DNA [140–142], IN inter- adopted to identify a linear peptide, LEDGF/p75325– acts strongly with the cellular transcriptional co-factor 530, that bound specifically to the IBD of LEDGF/p75. LEDGF/p75 [143]. LEDGF/p75 tethers the IN-DNA Based on this, small CPs (CP64 and CP65) inhibitors complex to the host chromatin, where the final integra- of the IN–LEDGF/p75 interaction (IC for CP64 tion steps take place [140,144–151]. The IN-LEDGF/p75 is 35.88 μM and IC for CP65 is 59.89 μM) were is a crucial interaction in the replication cycle, making developed. These peptides inhibited HIV replica-it as a fundamental target for anti-HIV drug design.
tion in different cell lines without displaying toxicity The structure of IN CCD in complex with the (Table 2C) [158]. Saturation transfer difference (STD) LEDGF/p75 IN binding domain (IBD) shows a pseudo two-fold symmetry where an IN CCD dimer Key termsbinds two LEDGF/p75 IBD at either side (PDB: 2BJ4, Alanine scan: Screening technique for determining of Figure 5B) [152]. The IBD interacts with IN via two loops. the contribution of specific residues to the function and Our lab rationally designed peptides based on these interactions of a protein. Each residue is sequential y replaced by alanine and the function/interaction of the loops and shorter variants (LEDGF/p75361–370, LEDGF/ mutant peptide/protein is compared with the parent p75402–411) [69]. All of these bound IN with micromolar peptide. Loss of function/interaction means that the affinities and inhibited the in vitro enzymatic activities original residue was important for the binding/activity. Alanine is used since it is the simplest chiral residue and both in presence and absence of LEDGF/p75 (Table 2B). thus mimic a loss of a side chain without a conformational In addition, these peptides inhibited the integration of change or an introduction of a new function.
viral cDNA and HIV-1 replication in infected cells, by Cyclic peptides: Cyclization improves the pharmacological shifting the IN oligomerization equilibrium toward a properties of peptides. They are conformational y rigid, stable tetramer in the cytosol. Further studies including resistant to protease degradation and in many cases have homology modeling, alanine scan and NMR analysis improved affinity and specificity as well as cell penetration revealed that all the residues of LEDGF/p75361–370 and properties.
future science group Review Chandra, Maes & Friedler NMR confirmed that the residues in CP64 strongly prevent PR-mediated cleavage at specific Gag sites and bound to LEDGF/p75 and not to HIV-1 IN.
also binds CA to prevent core formation [175].
Stapled peptides that target IN-mediated The interaction between Gag p6 & human integration & the IN-LEDGF/p75 interaction Two-domain crystal structures of IN show that the HIV-1 p6 is a Gag cleavage product that plays an two monomers of dimeric IN are tethered via strong important role in regulating capsid processing, facili-helix-helix (α1:α5′ and α5:α1′) interactions [159,160]. tating virus budding and incorporation of the viral Using the ‘sequence-walking' strategy, two potent IN accessory protein R (Vpr) into virions. These pro-inhibitors termed NL6 and NL9 [161] were revealed. cesses require interactions between the human tumor NL6 has an α-helical structure and is part of the α1 susceptibility gene 101 (Tsg101) protein and the CTD helical domain. A series of hydrocarbon stapled pep- of p6 [176]. Tsg101 is a part of the endosomal sorting tides derived from NL6 (NLH2-NLH16, NLX1, complex required for transport-I (ESCRT-I), which NLX2) enhanced interfacial interaction and cell- assists the ubiquitylation of Gag and facilitates viral permeability compared with the parent NL6 peptide assembly and budding [177–180]. Successful HIV-1 bud-through stabilization of the α1 domain [162] as con- ding requires an interaction between the tetrapeptide firmed by CD studies. Increasing the α-helical content PTAP, derived from residues 3–6 in p6, with the also increased the IN inhibitory activity at the 3′-pro- ubiquitin E2 variant (UEV) domain of Tsg101 (PDB: cessing step, inhibition of the strand transfer reaction 3OBU, 3OBX, Figure 6). Blocking this interaction and the IN-LEDGF/p75 interaction, cytoprotective inhibits virion formation [181–183].
activity (EC ), cell death activity (CC ) and thera- Peptides containing the PTAP motif are potential peutic index (ratio of CC to EC ). Combining pairs inhibitors of the interaction between Gag p6 and of α-helical peptides effectively inhibited IN catalytic Tsg101. A peptide derived from p65–13 bound Tsg101 activities. The most active pair was unstapled NLH5 (Table 3A) [184]. NMR studies showed that the peptide and stapled NLH6 (IC values of 9 ±1 μM for 3'-pro- bound to Tsg101 in a groove that interacts with the cessing and 6 ±1 μM for strand transfer [155]). The PTAP residues with K = 3 μM [177,183]. The struc- pairs were designed with a covalent hydrocarbon staple ture showed that binding of E2 ubiquitin-conjugating spanning i and i + 4 residues that did not show inhibi- enzymes to UEV domain of Tsg101 was hampered tion in the alanine scan [163,164]. Most of the stapled upon PTAP binding (Figure 6). Structure activity rela-peptide pairs inhibit the IN-LEDGF/p75 interaction. tionship (SAR) studies of this peptide, which included Six peptides (NLH2, NLH3, NLH5, NLH6, NLH15, conversion to P3 polycyclic oxime derivatives in the NLH16) inhibited HIV-1 replication in MT-4 cells. PTAP domain, improved binding to Tsg101 by 15- to Fluorescein-tagged NLH6 (termed NLX-1) pen- etrated cells and inhibited the target IN. MT-4 cells To develop effective competitive inhibitors, a tech- showed significant cellular uptake of NLX-1, which nique for genetically selecting CPs that inhibit specifi-was localized mainly to the cytoplasm with minimum cally the p6 Gag-Tsg101 interaction was used [187]. This distribution to the nucleus. The cell-permeability and technique, called SICLOPPS (split intein-mediated cir-enhanced potency of the stapled peptides makes them cular ligation of peptides and proteins), allowed iden-lead IN inhibitors.
tification of new CPs that specifically blocked the p6 Gag-Tsg101 interaction and consequently inhibited HIV Mapping HIV-1 Gag & host cellular proteins
replication. After several rounds of screening, the selected CPs had no resemblance to the original sequence of the HIV-1 Gag is a viral polyprotein expressed during the interacting sites in either p6 Gag or Tsg101. Of these, late phase of the replication cycle. Cleavage of Gag by the CP11 inhibited the formation of virus-like particles viral PR produces the structural proteins of the mature (VLP) in cultured cells with IC of 7μM and showed virion: the matrix (MA), capsid (CA) and nucleocapsid better stability compared with the linear p65–13.
(NC) proteins. MA is the N-terminus of Gag, followed by a CTD termed p6 and two spacer regions that sepa- The p6 Gag & cyclophilin interaction rate CA from NC and NC from p6. The Gag products Another partner of p6 Gag is the cellular cyclophilin take part in viral self-assembly and release of virions A (CypA) protein, which acts as a prolyl isomerase from the infected cells, thus making it critical for viral (PPIases). CypA also acts as a molecular chaperone particle morphogenesis and replication within the liv- and assists protein folding, assembly and transporta- ing cells [165–174]. The maturation inhibitor Bevirimat is tion processes. CypA is incorporated into newly bud-currently in clinical trials. It targets the Gag protein to ding particles of HIV-1 and thus can be considered as Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review a key target in future antiretroviral therapy [188]. p6 contains a relatively high content of proline residues, at positions 5, 7, 10, 11, 24, 30, 37 and 49. Proline cis/trans isomerism was observed for all these proline resi-dues and more than 40% of all p6 Gag proteins show at least one proline in cis-orientation. 2D proton NMR of full length p6 Gag or p6 Gag-derived peptides with CypA revealed that it interacts with all proline residues of p6 Gag through a prolyl-peptidyl cis-trans isomerase (PPIase).
The modulation of HIV-1 p6 function by CypA was explored by the synthesis of full length p6 and several Figure 6. Peptides derived from Tsg101 UEV. (A) Crystal
p6 fragments (p61–14, p61–21, p623–32, p632–42, p643–52 structure of the Tsg101 UEV domain (light brown) in and p623–52) and by using NMR and Surface Plasmon complex with a HIV-1 Gag P7A mutant p6 5–13 PSAP Resonance (SPR) (Table 3A) [188]. Catalytic amount peptide (green) (PDB:30BX) [285]. (B) Crystal structure
of CypA is sufficient to interact with all the proline of the Tsg101 UEV domain (blue) in complex with a residues of p61–52 (molar ratio 1: 283; Table 3A) and HIV-1 PTAP (5–13) peptide (red) (PDB:30BU) [285]. For color images please see online www.future-science.
hence PPIases activity in vitro. However, there was low com/doi/full/10.4155/FMC.15.46 affinity binding of CypA to p6 fragments compared with binding to full-length p6. Another important peptides have a higher resistance to degradation by inhibitor of CypA is cyclosporine A which was found proteases (Table 3B) [197,198,201–205].
to suppress both the production and the release of new virions [189,190].
The Tat-p53 interactionThe cellular tumor suppressor p53 is a homotetra- Interactions of HIV-1 Tat
meric transcription factor that induces cell cycle The HIV-1 trans-activator of transcription (Tat) arrest or apoptosis upon oncogenic stress [206]. NMR protein is a small viral auxiliary protein that con- and x-ray crystallography revealed that the p53 tet- tains 101 or 86 residues, depending on the HIV ramerization domain (p53 Tet; residues 326–355) strain [191,192]. The Tat protein can be divided into six has a dimer of dimers structure [207,208]. Depending regions: an acidic region (residues 2–11), a cysteine- on its concentration, p53 Tet exists in equilibrium rich domain (residues 22–37), the hydrophobic core among different oligomeric forms [209,210]. p53 inhib-(residues 38–46), a basic region (residues 47–57), its Tat-mediated LTR transcription [211]. The viral the glutamine-rich domain (residues 58–72) and Tat binds p53 Tet as was shown by yeast two-hybrid the RGD motif (residues 72–86) [193,194]. The basic system [209,212]. The CTD of p53 (residues 341–355) region of Tat binds to the negatively charged mRNA interacts specifically with the Tat residues 49–57 in in the Tat-activation region (TAR) [195,196]. The bind- the arginine-rich motif (ARM) [212]. Tat 73–86 can ing of Tat to TAR promotes a prolongation of the bind p53 with the assistance of cellular proteins such transcription due to conformational change of the as NF-κB and CBP/p300, as observed by in vivo TAR during binding of host cell kinases that phos- phorylate the RNA polymerase II complex. The six To quantitatively understand the molecular basis of Arginine residues in Tat47–57 are crucial for Tat-TAR Tat-p53 interaction during HIV-1 replication cycle, recognition [197–200].
our laboratory synthesized Tat-derived peptides (Tat1– The peptide Tat47–57 specifically disrupted the TAR- 35 and Tat47–57) and studied their binding to the p53 RNA recognition by blocking the production of viral tetramerization domain (Table 3C) [214]. The binding transcript and also interrupted the formation of two between p53 Tet and Tat47–57 is purely cooperative and cellular cofactors, cyclin T1 and its cognate kinase is temperature-dependent. NMR studies revealed that CDK9, responsible for transcriptional elongation E343 and E349 from p53 Tet are the major Tat47–57 from the viral long terminal repeat (LTR) [197,198,201– binding residues. The binding mechanism involves 205]. Increasing the number of Arginine residues on electrostatic interactions [214].
the hairpin scaffold of Tat-derived peptides dra-matically decreased the specificity for binding the The interaction of HIV-Vif with the host TAR-RNA. In contrast, fewer Arginine residues in cellular protein APOBEC3Ga Tat-derived peptide of the same length increased The HIV-1 virion infectivity factor (Vif) is required the TAR-RNA binding specificity. Arginine-rich Tat for the virus replication [215,216]. Vif counteracts A3G future science group Review Chandra, Maes & Friedler by targeting it for proteosomal degradation and by interaction between Vif and A3G and thus their inhi-direct inhibition of its enzymatic activity (PDB: bition may rescue the antiviral activity of A3G and 3IR2, Figure 4B) [217]. Both activities involve a direct inhibit HIV-1 propagation [218–220]. Vif binding to Table 3. Peptides derived from interactions between viral (Gag, Tat, Vif and Vpr) and host proteins.
(A) Interaction between MA and TCR
Gag p6 and Tsg101 interaction p6 5–13 PSAP peptides
p6 Gag and CypA interaction p6-UEV interaction (B) Interactions of Tat with host proteins
Tat derived peptides
(C) The Tat-p53 interaction
Tat derived peptides
P53 derived peptides p53 326–355 R342A p53 326–355 L344P p53 326–355 L344A p53 326–355 E346A †AghTat is (S)-2-Amino-6-guanidinohexanoic acid and AgbTat is (S)-2-Amino-4-guanidinobutyric acid. These are non natural amino acids.
PPI: Protein–protein interaction.
Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review Table 3. Peptides derived from interactions between viral (Gag, Tat, Vif and Vpr) and host proteins (cont.).
(D) Interactions between Vif and host proteins
Vif and A3G interaction
Vif and Cullin5 interaction (E) Interactions of Vpr
Vpr and CypA interaction
Vpr 75–90 (R80A) Vpr 75–90 (R76Q V83I Vpr 75–90 (R76Q V83I †AghTat is (S)-2-Amino-6-guanidinohexanoic acid and AgbTat is (S)-2-Amino-4-guanidinobutyric acid. These are non natural amino acids.
PPI: Protein–protein interaction.
A3G results in polyubquitination and degradation of from three distinct regions in Vif: residues 8–45 from A3G by forming a E3 ubiquitin ligase complex consist- the NTD, residues 154–192 from the CTD contain- ing of ElonginB and C, Cullin5 and RING finger pro- ing the conserved motif 161PPLP164 and a central region tein 1 [221,222]. The mutation K128D in A3G abrogated between residues 83–99 [230,231]. The A3G-derived pep-the interaction with Vif [223,224].
tides A3G143–157, A3G211–235 and A3G263–277 bound full- The peptides Vif14–17, Vif 22–26, Vif40–44 and Vif69–72 length Vif and Vif-CTD. The peptide array experiment inhibited the A3G-Vif interactions (Table 3D) [225]. revealed that peptides A3G 31–52, A3G 166–180, A3G 211–225,
Deletion mutagenesis of A3G also showed that A3G 263–277 and A3G 331–367 also bound to Vif.
Vif54–124 and Vif105–156 peptides are critical for the
interaction [226]. An in vitro Vif-A3G binding assay The interactions of Vpr with host cellular
between GST-tagged Vif and His-tagged A3G and proteins
a Fluorescence Resonance Energy Transfer (FRET) The viral protein R (Vpr) is the only virion associated reg-
assay between GST-Vif and biotinylated A3G110–148 ulatory protein and is not a component of the virus poly-
confirmed their interactions (Table 3D) [227].
protein precursors. It assists the nuclear import of the pre- Mapping the Vif-A3G interaction by using peptide integration complex (PIC) in nondividing host cells [232]. arrays resulted in defining the precise binding interface Vpr is crucial for effective HIV-1 infection of target (Table 3D) [226–229]. A3G bound nine Vif-derived peptides CD4+ T cells and macrophages [233–235]. Vpr interacts future science group Review Chandra, Maes & Friedler with numerous cellular proteins in order to perform its adenine nucleotide translocator (ANT) [246,247]. The nuclear import and G cell cycle arrest functions.
VDAC and ANT interaction is based on permeability transition pore (PTP) as a result of dynamic multiprotein Interaction between Vpr & CypA complex formation at inner and outer mitochondrial One of the key Vpr interacting protein is cyclophilin A membrane contact sites.
(CypA) [236]. Cis-trans prolyl isomerization of the highly A TEAM-VP (Targeted to Endothelial Apoptogenic conserved proline residues in Vpr, such as Pro5, Pro10, Mitochondrio-active Vpr-derived Peptide) peptide was Pro14 and Pro35, is catalyzed by CypA. SPR experiments designed based on α β binding and endothelial apop- showed that the heptapeptide CypA 32–38(32RHF- togenic sequences derived from the mitochondria active PRIW38) mediates the binding between CypA and the portion of Vpr. TEAM-VP peptide is combined with a N-terminal region of Vpr [237]. P35A mutation disrupted tumor blood vessel RGD-like ‘homing' motif and a mito-the Vpr-CypA interaction. In the mutant peptide Vpr75–90 chondrial membranes permealization (MMP)-inducing (R80A), the replacement in the C-terminal region of Vpr sequence. It is composed of the cysteine mediated CP hampered the co-IP of Vpr with CypA [238,239].
sequence GGCRGDMFGC and a Vpr67–82 sequence The above observations together with the significant derivative (Table 3E). The cyclic core ‘GGCRGDMFGC' amount of CypA in the virion [240] led to the design of of TEAM-VP specifically bound to VDAC and ANT Vpr-based peptides to study the Vpr - CypA interaction and internalized into α β -expessing cells through its (Table 3E) [241,242]. SPR and ITC studies revealed the cyclic-RGD motif. [248].
strong binding affinities of C-terminal Vpr75–90 (K = 0.28 μM) and N-terminal Vpr30–40 (K = 1 μM) peptides. PART II: interactions between viral proteins
Other C-terminal Vpr peptides such as Vpr69–78, Vpr75–84, The Env–MA interactionVpr81–90 and Vpr87–96 interacted weakly with CypA. The The matrix protein p17 (MA) originates from the weakest binding response was observed for mutant pep- Gag precursor protein, p55gag [249]. It is N-terminally tides such as Vpr75–90 R80A (K = 7.5 μM) and Vpr75–90 myristylated and binds to the viral inner membrane or R76Q, V83I, R80A, T841 (K = 4.7 μM) as compared the inner leaflet of the plasma membrane (PM) of the with the wild type peptide. NMR studies revealed that infected cells [250]. MA is involved in nuclear import of the mutations did not influence the secondary structure the viral DNA [251]. A specific interaction between p17 of the C-terminal binding domain of Vpr.
and Env was revealed by the co-expression of Env pro-teins that influenced the assembly of Gag particles. The The interaction between Vpr & the WXXF motif membrane-proximal amino terminus of p17 in the Gag of host cell proteins precursor closely associates with the membrane in the The conserved WXXF motif of uracil-DNA-glycosylase mature particle indicating that p17 participates in the mediates the intracellular binding of Vpr with uracil specific Env incorporation into the viral particles [252].
DNA glycosylase. Many WXXF-including peptides Several p17 peptides (p17l-12, p1712–29, p1730–52, p1753– have domain-specific interactions with Vpr. The fusion 87, p1787–115 and p17115–132), derived from all the six parts of the WXXF dimer to the chloramphenicol acetyl trans- of p17, were synthesized (Table 4A) [253,254]. The anti-ferase (CAT) gene demonstrated that the WXXF dimer- genic epitopes was examined for anti-HIV-1 p17 anti- CAT construct induced CAT activity inside the virions body (p17 Ab) in the serum of an HIV1 carrier. p17l-12, through Vpr-dependent docking [243].
p1712–29, p1730–52 were highly recognized in the serum Phage display peptide screening predicted that more and led to inhibition of virus multiplication as tested than 90% peptides having consensus motif WXXF effi- using ELISA. The purified antibodies obtained from the ciently binds Vpr protein [243,244]. Similarly, Vpr binding patient using the p17-derivated peptide immunoaffinity peptides from GST-Vpr panning also revealed a WXXF columns confirmed that the reactivity of p1730–52Ab to consensus motif [245]. Nine peptides were found to bind p17 was the highest among the antibodies.
Vpr (Table 3E) [243].
The Gag–PR interaction The Vpr interaction with cell-surface α β in PR cleaves the Gag and Gag-Pol precursors into active endothelial cells viral proteins such as p1gag, p2gag, p6gag, p7gag, p17gag and Vpr targets mitochondrial membranes to trigger apopto- p24gag [45,255–257]. The cleavage of the Gag precursors is sis and cell death. The internalization of cyclic RGD in necessary for maturation and HIV-1 infectivity. p2gag is endothelial cells for cellular apoptosis is mediated by the an inherent suicidal inhibitor of PR due to its strong in cell surface receptors α β integrins. The Vpr induced vitro inhibition of the proteolytic cleavage of the recom- apoptotic cell death involves the interactions of Vpr with binant Gag precursor into functional structural units the voltage-dependent anion channel (VDAC) and the (p17gag and p24gag) [258]. After the viral maturation, p2gag Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review inhibits PR activity in released viral particles and thus essential for PR binding and blocking proteolysis blocks the autolysis of HIV-1 virions.
(Table 4B). Vif21–65 inhibited PR five times better than PR is one of the most common anti-HIV drug tar- full-length Vif. Vif-derived peptides such as Vif30–65 and gets and many FDA approved anti-HIV drugs are PR Vif78–98 specifically inhibited the Vif-PR interaction in inhibitors [86,97]. The nonapeptide (AEAMSQVTN) vitro and blocked the production of viruses in HIV-1-in-derived from the N-terminus of p2gag inhibited HIV-1 fected cells [262,263]. Vif88–98 inhibited PR dimerization. PR activity in vitro to prevent autolysis of the virion after Two PR-derived peptides PR1–9 and PR94–99 abrogated sequential processing and reorganization of the virion Vif function as an A3G neutralizer and inhibited Vif-PR core (Table 4B) [258]. Further SAR studies with p2gag binding in a dose-dependent manner [264]. This means revealed that alanine replacements (M4A and T8A) and that PR1–9 competed with PR for the Vif binding site.
deletion of Asn9 from the nonamer (AEAMSQVTN) decreased the PR inhibitory properties. However, the Vpr interactions with RT & INother mutated peptides did not have inhibitory activity.
RT, IN and Vpr are in close spatial proximity within the PIC, allowing them to interact with each other [265]. The Vif–PR interaction The interaction between RT and IN involves the single- Vif blocks the cleavage activity by directly interacting stranded viral RNA copied into integration-competent with PR. Vif stably blocks the premature activation of double-stranded DNA by RT, DNA polymerase and PR in cytoplasm, which is circumvented during particle ribonuclease H (RNaseH). Then the PIC is imported assembly [259]. The NTD of Vif (residues 1–96) inhibits to nucleus by IN and Vpr for integration [266]. RT and the PR cleavage in vitro and in bacteria. Both Vif and PR IN physically interact with each other and the full-are present in the mature virions [260]. Vif regulates PR in length Vpr and its isolated CTD can interfere with the the virion at the early stage of infection [260]. Several Vif- IN-mediated integration activity in vitro [267].
derived peptides inhibited PR-mediated cleavage of Gag A library of Vpr-derived peptides was screened for in vitro and during viral protein expression in peripheral their ability to bind directly to RT and IN in vitro and blood lymphocytes [261]. Vif1–38 and Vif 1–65 and Vif10–96 to inhibit their enzymatic activities (Table 4B) [265–267]. peptides were highly stable toward proteolysis. Vif21–65 is Dot-blot binding assay showed that the C-terminal Vpr Table 4. Peptides derived from interactions between viral proteins.
(A) Interaction between Env and MA
Env and MA
(B) Interactions of Protease
Protease and Gag
p2 gag pep# mutant1 p2 gag pep# mutant2 p2 gag pep# mutant3 p2 gag pep# mutant4 p2 gag pep# mutant5 PPI: Protein–protein interaction.
future science group Review Chandra, Maes & Friedler peptides (Vpr57–71 and Vpr61–75) efficiently bound RT and Fuzeon® (Enfuvirtide) was approved for clinical use IN. Molecular docking of Vpr57–71 into the 3D structure against HIV [84–86,268,269]. Current research in anti-of RT and of the two peptides Vpr33–47 and Vpr61–75 into HIV drug design is focused on stabilizing lead peptides the IN CCD were carried out to understand the bio- using different strategies such as cyclization, peptoids chemical effects such as steric hindrance and conforma- and more [268–273].
tional changes of the active sites. DNA polymerase as well Peptides serve as excellent starting points for the as RNase H activities of RT were significantly inhibited design of peptidomimetics and the development of new by Vpr57–71, Vpr65–79 and Vpr69–83 with IC values in the small molecule drug leads based on their sequences and range of 0.22–2 μM. DNA primer extension by RT was conformations. Currently, many of the FDA-approved also inhibited by Vpr53–67, Vpr57–71, Vpr61–75, Vpr65–79 and anti-HIV drugs in the clinic, such as Indinavir, Ritona-Vpr69–83. Vpr33–47, Vpr57–71, Vpr61–75 and Vpr65–79 were able vir, Saquinavir and Lopinavir are the result of gradual to abrogate IN strand transfer activity. The three peptides conversion from a peptide to a small molecule [269,274–Vpr57–71, Vpr61–75 and Vpr65–79 inhibited the 3'-end process- 278]. These small molecules are mostly peptidomimetic ing activity of IN whereas the disintegration was blocked hydroxyethylene or hydroxymethylamine HIV-1 pro-by Vpr33–47, Vpr69–83, Vpr57–71, Vpr61–75 and Vpr65–79.
tease inhibitors. Other types of small molecules such as ADS-J1, ADS-J2, XTT formazan, NB-2, NB-64, Conclusion & future perspective
AOP-RANTES, PSC-RANTES, Vicriviroc, Mara- In this review, we described the PPI in the HIV-1 rep- viroc and Aplaviroc are also the outcome of peptido- lication cycle that are targets for inhibition by peptides mimetic approaches. They act by targeting the HIV-1 and from which inhibitory peptides were derived. These entry through gp120, gp41, CCR5 and CXCR4. This PPI include both viral–cellular and viral–viral protein approach may be used in the future for other PPI as interactions. Most of the peptides reported are derived described above.
from viral–host PPI and not from viral–viral PPI, indi-cating that the host–viral interactions are more promis- Financial & competing interests disclosure ing drug targets. Current research is focused on devel- A Friedler is supported by a grant from the Israeli Science oping peptides libraries based on in vitro and in vivo Foundation (ISF) and by the Minerva Centre for Bio-Hybrid experiments that will be later modified into small mol- Complex Systems. The authors have no other relevant affilia- ecule inhibitor. The peptides are discovered using dif- tions or financial involvement with any organization or entity ferent approaches, and different assays were performed with a financial interest in or financial conflict with the subject to analyze their quantitative or qualitative binding to matter or materials discussed in the manuscript apart from viral proteins and their effect on HIV-1 infectivity.
those disclosed.
Peptides do not serve only as tools for studying No writing assistance was utilized in the production of this PPI, but have clinical use against HIV. The peptide, manuscript.
Executive summary • Protein–protein interactions (PPI) are essential in every step of the human immunodeficiency virus (HIV) replication cycle.
• Mapping the interactions between viral and host proteins, as well as between the viral proteins themselves, is a fundamental target for the design and development of new therapeutics.
• Peptides are excellent tools to study the mechanisms of PPI in HIV-1 replication cycle and for the development of anti-HIV-1 drug leads that modulate PPI.
• These peptides can be later developed into small molecules, which can be used as drugs.
Chisholm ST, Coaker G, Day B, Staskawicz BJ. Host- microbe interactions: shaping the evolution of the plant Papers of special note have been highlighted as: immune response. Cell 124(4), 803–814 (2006).
• of interest; •• of considerable interest König R, Zhou Y, Elleder D et al. Global analysis of Jones S, Thornton JM. Principles of protein-protein host-pathogen interactions that regulate early-stage HIV-1 interactions. Proc. Natl Acad. Sci. USA 93(1), 13–20 (1996).
replication. Cell 135(1), 49–60 (2014).
Larsen TA, Olson AJ, Goodsell DS. Morphology of protein– Dixon RA, Lamb CJ. Molecular communication in protein interfaces. Structure 6(4), 421–427 (2014).
interactions between plants and microbial pathogens. Annu. Trakselis MA, Alley SC, Ishmael FT. Identification and Rev. Plant Physiol. Plant Mol. Biol. 41(1), 339–367 (1990).
mapping of protein–protein interactions by a combination Shapira SD, Gat-Viks I, Shum BO V et al. A physical of cross-linking, cleavage, and proteomics. Bioconjug. Chem. and regulatory map of host-influenza interactions reveals 16(4), 741–750 (2005).
pathways in H1N1 infection. Cell 139(7), 1255–1267 (2009).
Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review Baker B, Zambryski P, Staskawicz B, Dinesh-Kumar SP. Passow H. Molecular aspects of band 3 protein-mediated Signaling in plant-microbe interactions. Science 276(5313), anion transport across the red blood cell membrane. In: 726–733 (1997).
Reviews of Physiology, Biochemistry and Pharmacology, Volume Gu W, Schneider JW, Condorelli G, Kaushal S, Mahdavi 103 SE - 2. Springer, Berlin Heidelberg, 61–203 (1986).
V, Nadal-Ginard B. Interaction of myogenic factors Jager S, Cimermancic P, Gulbahce N et al. Global landscape and the retinoblastoma protein mediates muscle cell of HIV-human protein complexes. Nature 481(7381), commitment and differentiation. Cell 72(3), 309–324 365–370 (2012).
•• Information database for human and HIV-1 protein
Zervos AS, Gyuris J, Brent R. Mxi1, a protein that specifically interacts with Max to bind Myc-Max Brass AL, Dykxhoorn DM, Benita Y et al. Identification recognition sites. Cell 72(2), 223–232 (1993).
of host proteins required for HIV infection through a Kolch W. Meaningful relationships: the regulation of the functional genomic screen. Science 319(5865), 921–926 Ras/Raf/MEK/ERK pathway by protein interactions. Biochem. J. 351(2), 289–305 (2000).
Evans P, Dampier W, Ungar L, Tozeren A. Prediction of Pawson T, Nash P. Protein–protein interactions define HIV-1 virus-host protein interactions using virus and host specificity in signal transduction. Genes Dev. 14(9), sequence motifs. BMC Med. Genomics 2(1), 27 (2009).
1027–1047 (2000).
Pinney JW, Dickerson JE, Fu W, Sanders-Beer BE, Ptak Moran MF, Koch CA, Anderson D et al. Src homology RG, Robertson DL. HIV–host interactions: a map of viral region 2 domains direct protein-protein interactions in perturbation of the host system. AIDS 23(5), 549–554 signal transduction. Proc. Natl Acad. Sci. USA 87(21), 8622–8626 (1990).
•• Comprehensive protein–protein interaction database
Imagawa M, Chiu R, Karin M. Transcription factor AP-2 mediates induction by two different signal-transduction Von Mering C, Krause R, Snel B et al. Comparative pathways: protein kinase C and cAMP. Cell 51(2), assessment of large-scale data sets of protein-protein 251–260 (1987).
interactions. Nature 417(6887), 399–403 (2002).
Fields S, Song O. A novel genetic system to detect protein– Maulik U, Bhattacharyya M, Mukhopadhyay A, protein interactions. Nature 340(6230), 245–246 (1989).
Bandyopadhyay S. Identifying the immunodeficiency Wenger RH. Cellular adaptation to hypoxia: O2-sensing gateway proteins in humans and their involvement in protein hydroxylases, hypoxia-inducible transcription microRNA regulation. Mol. Biosyst. 7(6), 1842–1851 (2011).
factors, and O2-regulated gene expression. FASEB J. Fu W, Sanders-Beer BE, Katz KS, Maglott DR, Pruitt KD, 16(10), 1151–1162 (2002).
Ptak RG. Human immunodeficiency virus type 1, human Wu JY, Maniatis T. Specific interactions between proteins protein interaction database at NCBI. Nucleic Acids Res. implicated in splice site selection and regulated alternative 37(Suppl. 1), D417–D422 (2009).
splicing. Cell 75(6), 1061–1070 (1993).
Moustafa N, Eldin A S, Kassim KS. Predicting HIV-1 Campbell S, Vogt VM. Self-assembly in vitro of purified human protein interactions using data mining without CA-NC proteins from Rous sarcoma virus and human information loss. I JMEIT. 1(1), 25–41 (2013).
immunodeficiency virus type 1. J. Virol. 69(10), 6487– Dyer MD, Murali TM, Sobral BW. The landscape of human 6497 (1995).
proteins interacting with viruses and other pathogens. PLoS Sandalon Z, Oppenheim A. Self-assembly and protein– Pathog. 4(2), e32 (2008).
protein interactions between the SV40 capsid proteins Gavin A-C, Aloy P, Grandi P et al. Proteome survey reveals produced in insect cells. Virology 237(2), 414–421 (1997).
modularity of the yeast cell machinery. Nature 440(7084), Ceres P, Zlotnick A. Weak protein–protein interactions are 631–636 (2006).
sufficient to drive assembly of hepatitis B virus capsids†. Hakes L, Pinney JW, Robertson DL, Lovell SC. Protein- Biochemistry 41(39), 11525–11531 (2002).
protein interaction networks and biology -what's the Nielsen AL, Oulad-Abdelghani M, Ortiz JA, Remboutsika connection? Nat. Biotech. 26(1), 69–72 (2008).
E, Chambon P, Losson R. Heterochromatin formation in Fahey M, Bennett M, Mahon C et al. GPS-Prot: a web- mammalian cells: interaction between histones and HP1 based visualization platform for integrating host-pathogen proteins. Mol. Cell 7(4), 729–739 (2001).
interaction data. BMC Bioinformatics 12(1), 298 (2011).
Murzina N, Verreault A, Laue E, Stillman B. Budayeva H, Cristea I. A mass spectrometry view of Heterochromatin dynamics in mouse cells: interaction stable and transient protein interactions [Internet]. In: between chromatin assembly factor 1 and HP1 proteins. Advancements of Mass Spectrometry in Biomedical Research Mol. Cell 4(4), 529–540 (1999).
SE - 11. Woods AG, Darie CC (Eds.). Springer International Blobel G, Dobberstein B. Transfer of proteins across Publishing, 263–282 (2014).
membranes. I. Presence of proteolytically processed and Li S. Proteomics Defines Protein Interaction Network of unprocessed nascent immunoglobulin light chains on Signaling Pathways. In: Bioinformatics of Human Proteomics membrane-bound ribosomes of murine myeloma. J. Cell SE - 2. Wang X (Ed.). Springer Netherlands, 17–38 (2013).
Biol. 67(3), 835–851 (1975).
future science group Review Chandra, Maes & Friedler Morris JH, Knudsen GM, Verschueren E et al. Affinity Katzen F, Chang G, Kudlicki W. The past, present and purification–mass spectrometry and network analysis to future of cell-free protein synthesis. Trends Biotechnol. understand protein-protein interactions. Nat. Protoc. 9(11), 23(3), 150–156 (2005).
2539–2554 (2014).
Zhu H, Snyder M. Protein chip technology. Curr. Opin. Emig-Agius D, Olivieri K, Pache L et al. An integrated Chem. Biol. 7(1), 55–63 (2003).
map of HIV-human protein complexes that facilitate viral Roberts MJ, Bentley MD, Harris JM. Chemistry for peptide infection. PLoS ONE 9(5), e96687 (2014).
and protein PEGylation. Adv. Drug Deliv. Rev. 54(4), Ptak RG, Fu W, Sanders-Beer BE et al. Cataloguing the 459–476 (2002).
HIV type 1 human protein interaction network. AIDS Res. Engelhard VH, Altrich-Vanlith M, Ostankovitch M, Zarling Hum. Retroviruses 24(12), 1497–1502 (2008).
AL. Post-translational modifications of naturally processed Wheeler DL, Barrett T, Benson DA et al. Database MHC-binding epitopes. Curr. Opin. Immunol. 18(1), 92–97 resources of the national center for biotechnology information. Nucleic Acids Res. 35(Suppl. 1), D5–D12 Chandra K, Roy TK, Naoum JN, Gilon C, Gerber RB, Friedler A. A highly efficient in situ N-acetylation approach Van Dijk D, Ertaylan G, Boucher C, Sloot P. Identifying for solid phase synthesis. Org. Biomol. Chem. 12(12), potential survival strategies of HIV-1 through virus-host 1879–1884 (2014).
protein interaction networks. BMC Syst. Biol. 4(1), 96 Kerppola TK. Visualization of molecular interactions by fluorescence complementation. Nat. Rev. Mol. Cell Biol. 7(6), MacPherson JI, Dickerson JE, Pinney JW, Robertson DL. 449–456 (2006).
Patterns of HIV-1 protein interaction identify perturbed Dieterich DC, Lee JJ, Link AJ, Graumann J, Tirrell DA, host-cellular subsystems. PLoS Comput. Biol. 6(7), Schuman EM. Labeling, detection and identification of e1000863 (2010).
newly synthesized proteomes with bioorthogonal non- Frankel AD, Young JAT. HIV-1: fifteen proteins and an canonical amino-acid tagging. Nat. Protoc. 2(3), 532–540 RNA. Annu. Rev. Biochem. 67(1), 1–25 (1998).
Tavassoli A. Targeting the protein-protein interactions Lee LC, Hunter JJ, Mujeeb A, Turck C, Parslow TG. of the HIV lifecycle. Chem. Soc. Rev. 40(3), 1337–1346 Evidence for α-helical conformation of an essential n-terminal region in the human Bcl2 protein. J. Biol. Chem. Malim MH, Emerman M. HIV-1 accessory proteins– 271(38), 23284–23288 (1996).
ensuring viral survival in a hostile environment. Cell Host Sattler M, Liang H, Nettesheim D et al. Structure of Bcl- Microbe 3(6), 388–398 (2008).
xL-Bak peptide complex: recognition between regulators of Sakai K, Dimas J, Lenardo MJ. The Vif and Vpr accessory apoptosis. Science 275(5302), 983–986 (1997).
proteins independently cause HIV-1-induced T cell Eichler J. Peptides as protein binding site mimetics. Curr. cytopathicity and cell cycle arrest. Proc. Natl Acad. Sci. USA Opin. Chem. Biol. 12(6), 707–713 (2008).
103(9), 3369–3374 (2006).
Walensky LD, Kung AL, Escher I et al. Activation of Taylor BS, Sobieszczyk ME, McCutchan FE, Hammer SM. apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science The challenge of HIV-1 subtype diversity. N. Engl. J. Med. 305(5689), 1466–1470 (2004).
358(15), 1590–1602 (2008).
Murray JK, Gellman SH. Targeting protein–protein Rappaport M, Kogan J. HIV-1 accessory protein Vpr: interactions: lessons from p53/MDM2. Pept. Sci. 88(5), relevance in the pathogenesis of HIV and potential for 657–686 (2007).
therapeutic intervention. Retrovirology 8, 25 (2011).
Phan J, Li Z, Kasprzak A et al. Structure-based design of Tong AHY, Drees B, Nardelli G et al. A combined high affinity peptides inhibiting the interaction of p53 with experimental and computational strategy to define protein MDM2 and MDMX. J. Biol. Chem. 285(3), 2174–2183 interaction networks for peptide recognition modules. Science 295(5553), 321–324 (2002).
Pazgier M, Liu M, Zou G et al. Structural basis for high- Benyamini H, Friedler A. Using peptides to study protein– affinity peptide inhibition of p53 interactions with MDM2 protein interactions. Future Med. Chem. 2(6), 989–1003 and MDMX. Proc. Natl Acad. Sci. USA 106(12), 4665–4670 •• Review about the use of peptides to study protein–protein
Hayouka Z, Rosenbluh J, Levin A et al. Inhibiting HIV-1 integrase by shifting its oligomerization equilibrium. Proc. Natl Acad. Sci. USA 104(20), 8316–8321 (2007).
Tolsma SS, Volpert O V, Good DJ, Frazier WA, Polverini PJ, Bouck N. Peptides derived from two separate domains of Lawrence SH, Ramirez UD, Tang L et al. Shape shifting the matrix protein thrombospondin-1 have anti-angiogenic leads to small-molecule allosteric drug discovery. Chem. Biol. activity. J. Cell Biol. 122(2), 497–511 (1993).
15(6), 586–596 (2008).
Srivastava P. Interaction of heat shock proteins with Freed E. HIV-1 Replication. Somat. Cell Mol. Genet. peptides and antigen presenting cells: chaperoning of 26(1–6), 13–33 (2001).
the innate and adaptive immune responses. Annu. Rev. Freed EO. HIV-1 and the host cell: an intimate association. Immunol. 20(1), 395–425 (2002).
Trends Microbiol. 12(4), 170–177 (2004).
Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review Freed EO, Martin MA. The role of human immunodeficiency Brower ET, Schön A, Klein JC, Freire E. Binding virus type 1 envelope glycoproteins in virus infection. J. Biol. thermodynamics of the N-terminal peptide of the CCR5 Chem. 270(41), 23883–23886 (1995).
coreceptor to HIV-1 envelope glycoprotein gp120†. Kwong PD, Wyatt R, Robinson J, Sweet RW, Sodroski J, Biochemistry 48(4), 779–785 (2009).
Hendrickson WA. Structure of an HIV gp120 envelope Davis CB, Dikic I, Unutmaz D et al. Signal transduction due glycoprotein in complex with the CD4 receptor and a to HIV-1 envelope interactions with chemokine receptors neutralizing human antibody. Nature 393(6686), 648–659 CXCR4 or CCR5. J. Exp. Med. 186(10), 1793–1798 (1997).
Rizzuto CD, Wyatt R, Hernández-Ramos N et al. A Moore JP, Binley J. HIV: envelope's letters boxed into shape. conserved HIV gp120 glycoprotein structure involved in Nature 393(6686), 630–631 (1998).
chemokine receptor binding. Science 280(5371), 1949–1953 Wyatt R, Kwong PD, Desjardins E et al. The antigenic structure of the HIV gp120 envelope glycoprotein. Nature Möbius K, Dürr R, Haußner C, Dietrich U, Eichler J. A 393(6686), 705–711 (1998).
functionally selective synthetic mimic of the HIV-1 co- Pancera M, Majeed S, Ban Y-EA et al. Structure of HIV-1 receptor CXCR4. Chem. Eur. J. 18(27), 8292–8295 (2012).
gp120 with gp41-interactive region reveals layered envelope Gross A, Möbius K, Haußner C, Donhauser N, Schmidt B, architecture and basis of conformational mobility. Proc. Natl Eichler J. Exploring converse molecular mechanisms of anti- Acad. Sci. USA 107(3), 1166–1171 (2010).
HIV-1 antibodies using a synthetic CXCR4 mimic. Bioorg. Eckert DM, Kim PS. Mechanisms of viral membrane fusion Med. Chem. Lett. 22(19), 6099–6102 (2012).
and its inhibition. Ann. Rev. Biochem. 70(1), 777–810 (2001).
Zennou V, Petit C, Guetard D, Nerhbass U, Montagnier L, Deng H, Liu R, Ellmeier W et al. Identification of a major Charneau P. HIV-1 genome nuclear import is mediated by a co-receptor for primary isolates of HIV-1. Nature 381(6584), central DNA flap. Cell 101(2), 173–185 (2000).
661–666 (1996).
Kohlstaedt LA, Wang J, Friedman JM, Rice PA, Steitz Dragic T, Litwin V, Allaway GP et al. HIV-1 entry into TA. Crystal structure at 3.5 A resolution of HIV-1 reverse CD4+ cells is mediated by the chemokine receptor CC- transcriptase complexed with an inhibitor. Science 256(5065), CKR-5. Nature 381(6584), 667–673 (1996).
1783–1790 (1992).
He J, Chen Y, Farzan M et al. CCR3 and CCR5 are co- Wang J, Smerdon SJ, Jäger J et al. Structural basis of receptors for HIV-1 infection of microglia. Nature 385(6617), asymmetry in the human immunodeficiency virus type 1 645–649 (1997).
reverse transcriptase heterodimer. Proc. Natl Acad. Sci. USA 91(15), 7242–7246 (1994).
Chan DC, Fass D, Berger JM, Kim PS. Core structure of gp41 from the HIV Envelope Glycoprotein. Cell 89(2), Jochmans D. Novel HIV-1 reverse transcriptase inhibitors. 263–273 (2014).
Vir. Res. 134(1–2), 171–185 (2008).
Marsh M, Helenius A. Virus entry: open sesame. Cell 124(4), •• Overview of reverse trascriptase inhibitors with the
729–740 (2006).
strategy for their development.
Doranz BJ, Rucker J, Yi Y et al. A dual-tropic primary 100 Iwatani Y, Chan DSB, Wang F et al. Deaminase-independent HIV-1 isolate that uses fusin and the β-chemokine receptors inhibition of HIV-1 reverse transcription by APOBEC3G. CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell 85(7), Nucleic Acids Res. 35(21), 7096–7108 (2007).
1149–1158 (1996).
101 Wang X, Ao Z, Chen L, Kobinger G, Peng J, Yao X. The Gross A, Möbius K, Haußner C, Donhauser N, Schmidt B, cellular antiviral protein APOBEC3G interacts with HIV-1 Eichler J. Mimicking protein-protein interactions through reverse transcriptase and inhibits its function during viral peptide-peptide interactions: HIV-1 gp120 and CXCR4. replication. J. Virol. 86(7), 3777–3786 (2012).
Front. Immunol. 4(257), 1–11 (2013).
102 Stainforth DA, Aina T, Christensen C et al. Uncertainty Esté JA, Telenti A. HIV entry inhibitors. Lancet 370(9581), in predictions of the climate response to rising levels of 81–88 (2015).
greenhouse gases. Nature 433(7024), 403–406 (2005).
Rao BS, Gupta KK, Kumari S, Gupta A, Pujitha K. 103 Yu X, Yu Y, Liu B et al. Induction of APOBEC3G Conserved HIV wide spectrum antipeptides-a hope for HIV ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF treatment. Adv. Tech. Biol. Med. 1(1), 1–9 (2013).
complex. Science 302(5647), 1056–1060 (2003).
Teissier E, Penin F, Pécheur E-I. Targeting cell entry of 104 Mangeat B, Turelli P, Caron G, Friedli M, Perrin L, Trono enveloped viruses as an antiviral strategy. Molecules 16(1), D. Broad antiretroviral defence by human APOBEC3G 221–250 (2010).
through lethal editing of nascent reverse transcripts. Nature 424(6944), 99–103 (2003).
Pascual R, Contreras M, Fedorov A, Prieto M, Villalaín J. Interaction of a peptide derived from the N-heptad repeat 105 Miyagi E, Opi S, Takeuchi H et al. Enzymatically region of gp41 Env ectodomain with model membranes. active APOBEC3G is required for efficient inhibition of modulation of phospholipid phase behavior. Biochemistry human immunodeficiency virus type 1. J. Virol. 81(24), 44(43), 14275–14288 (2005).
Huang C, Lam SN, Acharya P et al. Structures of the CCR5 106 Li X-Y, Guo F, Zhang L, Kleiman L, Cen S. APOBEC3G N terminus and of a tyrosine-sulfated antibody with HIV-1 inhibits DNA strand transfer during HIV-1 reverse gp120 and CD4. Science 317(5846), 1930–1934 (2007).
transcription. J. Biol. Chem. 282(44), 32065–32074 (2007).
future science group Review Chandra, Maes & Friedler 107 Guo F, Cen S, Niu M, Saadatmand J, Kleiman L. Inhibition 122 Leh H, Brodin P, Bischerour J et al. Determinants of of -primed reverse transcription by human APOBEC3G Mg2+-dependent activities of recombinant human during human immunodeficiency virus type 1 replication. immunodeficiency virus type 1 integrase†. Biochemistry J. Virol. 80(23), 11710–11722 (2006).
39(31), 9285–9294 (2000).
108 Guo F, Saadatmand J, Niu M, Kleiman L. Roles of Gag 123 Li M, Craigie R. Processing of viral DNA ends channels and NCp7 in facilitating annealing to viral RNA in human the HIV-1 Integration Reaction to Concerted Integration. immunodeficiency virus type 1. J. Virol. 83(16), 8099–8107 J. Biol. Chem. 280(32), 29334–29339 (2005).
124 Faure A, Calmels C, Desjobert C et al. HIV-1 integrase 109 Druillennec S, Dong CZ, Escaich S et al. A mimic of HIV-1 crosslinked oligomers are active in vitro. Nucleic Acids Res. nucleocapsid protein impairs reverse transcription and 33(3), 977–986 (2005).
displays antiviral activity. Proc. Natl Acad. Sci. USA 96(9), 125 Chaurushiya MS, Weitzman MD. Viral manipulation of 4886–4891 (1999).
DNA repair and cell cycle checkpoints. DNA Repair 8(9), 110 De Rocquigny H, Gabus C, Vincent A, Fournié-Zaluski 1166–1176 (2009).
MC, Roques B, Darlix JL. Viral RNA annealing activities of 126 Lewinski MK, Bushman FD. Retroviral DNA integration– human immunodeficiency virus type 1 nucleocapsid protein mechanism and consequences. Adv. Genet. 55, 147–181 require only peptide domains outside the zinc fingers. Proc. Natl Acad. Sci. USA 89(14), 6472–6476 (1992).
127 Parissi V, Caumont A, de Soultrait VR et al. The lethal 111 Craigie R. HIV Integrase, a Brief Overview from Chemistry phenotype observed after HIV-1 integrase expression in yeast to Therapeutics. J. Biol. Chem. 276(26), 23213–23216 cells is related to DNA repair and recombination events. Gene 322(0), 157–168 (2003).
112 Delelis O, Carayon K, Saib A, Deprez E, Mouscadet J-F. 128 Oyebisi Jegede, John Babu, Roberto di Santo, Damian J, Integrase and integration: biochemical activities of HIV-1 McColl JW, MEQ-M. HIV type 1 integrase inhibitors: from integrase. Retrovirology 5(1), 114 (2008).
basic research to clinical implications. AIDS Rev. 10(3), 113 Schröder ARW, Shinn P, Chen H, Berry C, Ecker JR, 172–189 (2008).
Bushman F. HIV-1 integration in the human genome favors 129 Freed CSA and EO. Anti-HIV-1 therapeutics: from FDA- active genes and local hotspots. Cell 110(4), 521–529 (2014).
approved drugs to hypothetical future targets. Mol. Interv. 114 Kalpana G V, Marmon S, Wang W, Crabtree GR, Goff Apr. 9(2), 70–74 (2009).
SP. Binding and stimulation of HIV-1 integrase by a 130 Maes M, Loyter A, Friedler A. Peptides that inhibit HIV-1 human homolog of yeast transcription factor SNF5. Science integrase by blocking its protein-protein interactions. FEBS J. 266(5193), 2002–2006 (1994).
279(16), 2795–2809 (2012).
115 Stevenson M, Stanwick T L, Dempsey M P, Lamonica Review about peptides that target the protein–protein
CA. HIV-1 replication is controlled at the level of T interactions of HIV-1 integrase.
cell activation and proviral integration. EMBO J. 9(5), 1551–1560 (1990).
131 Camarasa M-J, Velázquez S, San-Félix A, Pérez-Pérez M-J, Gago F. Dimerization inhibitors of HIV-1 reverse 116 Chen JC, Krucinski J, Miercke LJ et al. Crystal structure of transcriptase, protease and integrase: A single mode of the HIV-1 integrase catalytic core and C-terminal domains: inhibition for the three HIV enzymes? Antiviral. Res. a model for viral DNA binding. Proc. Natl Acad. Sci. USA 71(2–3), 260–267 (2006).
97(15), 8233–8238 (2000).
132 Soultrait VR d, Desjobert C, Tarrago-Litvak L. Peptides as 117 Dyda F, Hickman AB, Jenkins TM, Engelman A, Craigie new inhibitors of HIV-1 reverse transcriptase and integrase. R, Davies DR. Crystal structure of the catalytic domain Curr. Med. Chem. 10(18), 1765–1778 (2003).
of HIV-1 integrase: similarity to other polynucleotidyl transferases. Science 266(5193), 1981–1986 (1994).
133 Goldgur Y, Dyda F, Hickman AB, Jenkins TM, Craigie R, Davies DR. Three new structures of the core domain of HIV- 118 Marchand C, Johnson AA, Semenova E, Pommier Y. 1 integrase: an active site that binds magnesium. Proc. Natl Mechanisms and inhibition of HIV integration. Drug Acad. Sci. USA 95(16), 9150–9154 (1998).
Discov. Today Dis. Mech. 3(2), 253–260 (2006).
134 Neamati N, Sunder S, Pommier Y. Design and discovery HIV-1 integration and its role in replication.
of HIV-1 integrase inhibitors. Drug Discov. Today 2(11), 119 Guiot E, Carayon K, Delelis O et al. Relationship between 487–498 (1997).
the oligomeric status of HIV-1 integrase on DNA and 135 Sourgen F, Maroun RG, Frère V et al. A synthetic peptide enzymatic activity. J. Biol. Chem. 281(32), 22707–22719 from the human immunodeficiency virus Type-1 integrase exhibits coiled-coil properties and interferes with the in vitro 120 Van Maele B, Busschots K, Vandekerckhove L, Christ F, integration activity of the enzyme. Euro J. Biochem. 240(3), Debyser Z. Cellular co-factors of HIV-1 integration. Trends 765–773 (1996).
Biochem. Sci. 31(2), 98–105 (2014).
136 Maroun RG, Gayet S, Benleulmi MS et al. Peptide inhibitors 121 Levin A, Armon-Omer A, Rosenbluh J et al. Inhibition of of HIV-1 integrase dissociate the enzyme oligomers†. HIV-1 integrase nuclear import and replication by a peptide Biochemistry 40(46), 13840–13848 (2001).
bearing integrase putative nuclear localization signal. 137 Zhao L, O'Reilly MK, Shultz MD, Chmielewski J. Retrovirology 6(112), 1–16 (2009).
Interfacial peptide inhibitors of HIV-1 integrase activity and Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review dimerization. Bioorg. Med. Chem. Lett. 13(6), 1175–1177 152 Cherepanov P, Ambrosio ALB, Rahman S, Ellenberger T, Engelman A. Structural basis for the recognition between 138 Van Aerschot A. Oligonucleotides as antivirals: dream HIV-1 integrase and transcriptional coactivator p75. Proc. or realistic perspective? Antiviral Res. 71(2–3), 307–316 Natl Acad. Sci. USA 102(48), 17308–17313 (2005).
153 Levin A, Benyamini H, Hayouka Z, Friedler A, Loyter A. 139 Archakov AI, Govorun VM, Dubanov A V et al. Protein- Peptides that bind the HIV-1 integrase and modulate its protein interactions as a target for drugs in proteomics. enzymatic activity – kinetic studies and mode of action. Proteomics 3(4), 380–391 (2003).
FEBS J. 278(2), 316–330 (2011).
140 Emiliani S, Mousnier A, Busschots K et al. Integrase 154 Hayouka Z, Levin A, Maes M et al. Mechanism of action mutants defective for interaction with LEDGF/p75 are of the HIV-1 integrase inhibitory peptide LEDGF 361– impaired in chromosome tethering and HIV-1 replication. 370. Biochem. Biophys. Res. Commun. 394(2), 260–265 J. Biol. Chem. 280(27), 25517–25523 (2005).
141 Turlure F, Devroe E, Silver P A, Engelman A. Human 155 Hayouka Z, Hurevich M, Levin A et al. Cyclic peptide cell proteins and human immunodeficiency virus DNA inhibitors of HIV-1 integrase derived from the LEDGF/p75 integration. Front. Biosci. 9, 3187–3208 (2004).
protein. Bioorg. Med. Chem. 18(23), 8388–8395 (2010).
142 Al-Mawsawi LQ, Neamati N. Blocking interactions between 156 Maes M, Levin A, Hayouka Z, Shalev DE, Loyter A, HIV-1 integrase and cellular cofactors: an emerging anti- Friedler A. Peptide inhibitors of HIV-1 integrase: From retroviral strategy. Trends Pharmacol. Sci. 28, 526–535 mechanistic studies to improved lead compounds. Bioorg. Med. Chem. 17(22), 7635–7642 (2009).
143 Busschots K, Vercammen J, Emiliani S et al. The interaction 157 Al-Mawsawi LQ, Christ F, Dayam R, Debyser Z, Neamati of LEDGF/p75 with integrase is lentivirus-specific N. Inhibitory profile of a LEDGF/p75 peptide against HIV- and promotes DNA binding. J. Biol. Chem. 280(18), 1 integrase: Insight into integrase–DNA complex formation and catalysis. FEBS Lett. 582(10), 1425–1430 (2008).
144 Llano M, Vanegas M, Fregoso O et al. LEDGF/p75 158 Desimmie BA, Humbert M, Lescrinier E et al. Phage determines cellular trafficking of diverse lentiviral but not display-directed discovery of LEDGF/p75 binding cyclic murine oncoretroviral integrase proteins and is a component peptide inhibitors of HIV replication. Mol. Ther. 20(11), of functional lentiviral preintegration complexes. J. Virol. 2064–2075 (2012).
78(17), 9524–9537 (2004).
159 Wang J-Y, Ling H, Yang W, Craigie R. Structure of a 145 Cherepanov P, Maertens G, Proost P et al. HIV-1 integrase two-domain fragment of HIV-1 integrase: implications forms stable tetramers and associates with LEDGF/p75 for domain organization in the intact protein. EMBO J. protein in human cells. J. Biol. Chem. 278(1), 372–381 20(24), 7333–7343 (2001).
160 Dyda F, Hickman AB, Jenkins TM, Engelman A, Craigie 146 Vandegraaff N, Devroe E, Turlure F, Silver PA, Engelman R, Davies D. Crystal structure of the catalytic domain A. Biochemical and genetic analyses of integrase-interacting of HIV-1 integrase: similarity to other polynucleotidyl proteins lens epithelium-derived growth factor (LEDGF)/ transferases. Science 266, 1981–1986 (2004).
p75 and hepatoma-derived growth factor related protein 161 Chen H, Engelman A. Characterization of a replication- 2 (HRP2) in preintegration complex function and HIV-1 defective human immunodeficiency virus type 1 att site replication. Virology 346(2), 415–426 (2006).
mutant that is blocked after the 3′ processing step of 147 Ciuffi A, Llano M, Poeschla E et al. A role for LEDGF/ retroviral integration. J. Virol. 74(17), 8188–8193 (2000).
p75 in targeting HIV DNA integration. Nat. Med. 11(12), 162 Long Y-Q, Huang S-X, Zawahir Z et al. Design of cell- 1287–1289 (2005).
permeable stapled peptides as HIV-1 integrase inhibitors. 148 Llano M, Delgado S, Vanegas M, Poeschla EM. Lens J. Med. Chem. 56(13), 5601–5612 (2013).
epithelium-derived growth factor/p75 prevents proteasomal 163 Desjobert C, de Soultrait VR, Faure A et al. Identification degradation of HIV-1 integrase. J. Biol. Chem. 279(53), by phage display selection of a short peptide able to inhibit only the strand transfer reaction catalyzed by human 149 Cherepanov P, Sun Z-YJ, Rahman S, Maertens G, Wagner immunodeficiency virus type 1 integrase†. Biochemistry G, Engelman A. Solution structure of the HIV-1 integrase- 43(41), 13097–13105 (2004).
binding domain in LEDGF/p75. Nat. Struct. Mol. Biol. 164 Li H-Y, Zawahir Z, Song L-D, Long Y-Q, Neamati N. 12(6), 526–532 (2005).
Sequence-based design and discovery of peptide inhibitors 150 Llano M, Saenz DT, Meehan A et al. An essential role of HIV-1 integrase: insight into the binding mode of the for LEDGF/p75 in HIV integration. Science 314(5798), enzyme. J. Med. Chem. 49(15), 4477–4486 (2006).
461–464 (2006).
165 Freed EO. HIV-1 Gag proteins: diverse functions in the 151 Cherepanov P, Devroe E, Silver PA, Engelman A. virus life cycle. Virology 251(1), 1–15 (1998).
Identification of an evolutionarily conserved domain in 166 Saad JS, Miller J, Tai J, Kim A, Ghanam RH, Summers human lens epithelium-derived growth factor/transcriptional MF. Structural basis for targeting HIV-1 Gag proteins to co-activator p75 (LEDGF/p75) that binds HIV-1 integrase. the plasma membrane for virus assembly. Proc. Natl Acad. J. Biol. Chem. 279(47), 48883–48892 (2004).
Sci. USA 103(30), 11364–11369 (2006).
future science group Review Chandra, Maes & Friedler 167 Bennett RP, Nelle TD, Wills JW. Functional chimeras of the 183 Demirov DG, Ono A, Orenstein JM, Freed EO. Rous sarcoma virus and human immunodeficiency virus gag Overexpression of the N-terminal domain of TSG101 proteins. J. Virol. 67(11), 6487–6498 (1993).
inhibits HIV-1 budding by blocking late domain function. 168 Craven RC, Leure-duPree AE, Weldon RA, Wills JW. Proc. Natl Acad. Sci. USA 99(2), 955–960 (2002).
Genetic analysis of the major homology region of the Rous 184 Kim S-E, Liu F, Im YJ et al. Elucidation of new binding sarcoma virus Gag protein. J. Virol. 69(7), 4213–4227 interactions with the human Tsg101 protein using modified HIV-1 Gag-p6 derived peptide ligands. ACS Med. Chem. 169 Dawson L, Yu X-F. The role of nucleocapsid of HIV-1 in Lett. 2(5), 337–341 (2011).
virus assembly. Virology 251(1), 141–157 (1998).
185 Liu F, Stephen AG, Waheed AA et al. SAR by oxime- 170 Kenney SP, Lochmann TL, Schmid CL, Parent LJ. containing peptide libraries: application to Tsg101 ligand Intermolecular Interactions between retroviral Gag proteins optimization. ChemBioChem 9(12), 2000–2004 (2008).
in the nucleus. J. Virol. 82(2), 683–691 (2008).
186 Liu F, Stephen AG, Fisher RJ, Burke Jr. TR. Protected 171 Wills JW, Craven RC. Form, function, and use of retroviral aminooxyprolines for expedited library synthesis: application Gag proteins. AIDS 5(6) (1991).
to Tsg101-directed proline–oxime containing peptides. Bioorg. Med. Chem. Lett. 18(3), 1096–1101 (2008).
172 Martinez-Hackert E, Anikeeva N, Kalams SA, Walker BD, Hendrickson WA, Sykulev Y. Structural basis for degenerate 187 Tavassoli A, Lu Q, Gam J, Pan H, Benkovic SJ, Cohen SN. recognition of natural HIV peptide variants by cytotoxic Inhibition of HIV budding by a genetically selected cyclic lymphocytes. J. Biol. Chem. 281(29), 20205–20212 (2006).
peptide targeting the Gag–TSG101 interaction. ACS Chem. Biol. 3(12), 757–764 (2008).
173 Brander C, Hartman KE, Trocha AK et al. Lack of strong immune selection pressure by the immunodominant, Development of cyclic pepdtide that target HIV-1
HLA-A*0201-restricted cytotoxic T lymphocyte response in chronic human immunodeficiency virus-1 infection. J. Clin. 188 Solbak SMØ, Reksten TR, Röder R et al. HIV-1 p6– Investig. 101(11), 2559–2566 (1998).
Another viral interaction partner to the host cellular protein 174 Goulder PJR, Sewell AK, Lalloo DG et al. Patterns cyclophilin A. Biochim. Biophys. Acta 1824(4), 667–678 of immunodominance in HIV-1–specific cytotoxic T lymphocyte responses in two human histocompatibility 189 Hatziioannou T, Perez-Caballero D, Cowan S, Bieniasz leukocyte antigens (HLA)-identical siblings with PD. Cyclophilin interactions with incoming human HLA-A*0201 are influenced by epitope mutation. J. Exp. immunodeficiency virus type 1 capsids with opposing effects Med. 185(8), 1423–1433 (1997).
on infectivity in human cells. J. Virol. 79(1), 176–183 175 Li F, Goila-Gaur R, Salzwedel K et al. PA-457: a potent HIV inhibitor that disrupts core condensation by targeting a late 190 Franke EK, Luban J. Inhibition of HIV-1 replication by step in Gag processing. Proc. Natl Acad. Sci. USA 100(23), cyclosporine A or related compounds correlates with the ability to disrupt the Gag–cyclophilin a interaction. Virology 176 Huang M, Orenstein JM, Martin MA, Freed EO. p6Gag 222(1), 279–282 (1996).
is required for particle production from full-length human 191 Rana TM, Jeang K-T. Biochemical and functional immunodeficiency virus type 1 molecular clones expressing interactions between HIV-1 Tat protein and TAR RNA. protease. J. Virol. 69(11), 6810–6818 (1995).
Arch. Biochem. Biophys. 365(2), 175–185 (1999).
177 Pornillos O, Alam SL, Davis DR, Sundquist WI. Structure 192 Sodroski J, Patarca R, Rosen C, Wong-Staal F, Haseltine of the Tsg101 UEV domain in complex with the PTAP W. Location of the trans-activating region on the genome of motif of the HIV-1 p6 protein. Nat. Struct. Mol. Biol. 9(11), human T-cell lymphotropic virus type III. Science 229(4708), 812–817 (2002).
74–77 (1985).
178 Garrus JE, von Schwedler UK, Pornillos OW et al. Tsg101 193 Bayer P, Kraft M, Ejchart A, Westendorp M, Frank R, Rösch and the vacuolar protein sorting pathway are essential for P. Structural studies of HIV-1 Tat protein. J. Mol. Biol. HIV-1 budding. Cell 107(1), 55–65 (2014).
247(4), 529–535 (1995).
179 VerPlank L, Bouamr F, LaGrassa TJ et al. Tsg101, a 194 Churcher MJ, Lamont C, Hamy F et al. High affinity homologue of ubiquitin-conjugating (E2) enzymes, binds the binding of TAR RNA by the human immunodeficiency virus L domain in HIV type 1 Pr55Gag. Proc. Natl Acad. Sci. USA type-1 Tat protein requires base-pairs in the rna stem and 98(14), 7724–7729 (2001).
amino acid residues flanking the basic region. J. Mol. Biol. 180 Martin-Serrano J, Zang T, Bieniasz PD. HIV-1 and Ebola 230(1), 90–110 (1993).
virus encode small peptide motifs that recruit Tsg101 to sites 195 Long KS, Crothers DM. Interaction of Human of particle assembly to facilitate egress. Nat. Med. 7(12), Immunodeficiency Virus Type 1 Tat-derived peptides with 1313–1319 (2001).
TAR RNA. Biochemistry 34(27), 8885–8895 (1995).
181 Zhan P, Li W, Chen H, Liu X. Targeting protein-protein 196 Luo Y, Madore SJ, Parslow TG, Cullen BR, Peterlin interactions: a promising avenue of anti-HIV drug discovery. BM. Functional analysis of interactions between Tat Curr. Med. Chem. 17(29), 3393–3409 (2010). and the trans-activation response element of human 182 Freed EO. The HIV–TSG101 interface: recent advances in a immunodeficiency virus type 1 in cells. J. Virol. 67(9), budding field. Trends Microbiol. 11(2), 56–59 (2003).
5617–5622 (1993).
Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review 197 Cullen BR. Regulation of HIV-1 gene expression. FASEB J. immunodeficiency virus type 1. Proc. Natl Acad. Sci. USA 5(10), 2361–2368 (1991).
92(12), 5461–5464 (1995).
198 Calnan BJ, Biancalana S, Hudson D, Frankel AD. Analysis 213 Cullen R. Mechanism of action of regulatory proteins of Arginine-rich peptides from the HIV Tat protein reveals encoded by complex retroviruses. Microbiol. Rev. 56(3), unusual features of RNA-protein recognition. Genes Dev. 375–394 (1992).
5(2), 201–210 (1991).
214 Gabizon R, Mor M, Rosenberg MM et al. Using peptides 199 Ensoli B, Buonaguro L, Barillari G et al. Release, uptake, to study the interaction between the p53 tetramerization and effects of extracellular human immunodeficiency virus domain and HIV-1 tat. Peptide Sci. 90(2), 105–116 (2008).
type 1 Tat protein on cell growth and viral transactivation. 215 Fisher AG, Ensoli B, Ivanoff L et al. The sor gene of HIV-1 J. Virol. 67(1), 277–287 (1993).
is required for efficient virus transmission in vitro. Science 200 Stevens M, De Clercq E, Balzarini J. The regulation of 237(4817), 888–893 (1987).
HIV-1 transcription: molecular targets for chemotherapeutic 216 Gabuzda DH, Lawrence K, Langhoff E et al. Role of vif intervention. Med. Res. Rev. 26(5), 595–625 (2006).
in replication of human immunodeficiency virus type 1 in 201 Weeks KM, Ampe C, Schultz SC, Steitz TA, Crothers DM. CD4+ T lymphocytes. J. Virol. 66(11), 6489–6495 (1992).
Fragments of the HIV-1 Tat protein specifically bind TAR 217 Huthoff H, Malim MH. Cytidine deamination and RNA. Science 249(4974), 1281–1285 (1990).
resistance to retroviral infection: towards a structural 202 Cordingley MG, LaFemina RL, Callahan PL et al. Sequence- understanding of the APOBEC proteins. Virology 334(2), specific interaction of Tat protein and Tat peptides with 147–153 (2005).
the transactivation-responsive sequence element of human 218 Harris RS, Bishop KN, Sheehy AM et al. DNA deamination immunodeficiency virus type 1 in vitro. Proc. Natl Acad. Sci. mediates innate immunity to retroviral infection. Cell USA 87(22), 8985–8989 (1990).
113(6), 803–809 (2003).
203 Gelman MA, Richter S, Cao H, Umezawa N, Gellman SH, 219 Mariani R, Chen D, Schröfelbauer B et al. Species-specific Rana TM. Selective Binding of TAR RNA by a Tat-Derived exclusion of APOBEC3G from HIV-1 virions by Vif. Cell β-Peptide. Org. Lett. 5(20), 3563–3565 (2003).
114(1), 21–31 (2003).
204 Wender PA, Mitchell DJ, Pattabiraman K, Pelkey ET, 220 Zhang H, Yang B, Pomerantz RJ, Zhang C, Arunachalam Steinman L, Rothbard JB. The design, synthesis, and SC, Gao L. The cytidine deaminase CEM15 induces evaluation of molecules that enable or enhance cellular hypermutation in newly synthesized HIV-1 DNA. Nature uptake: peptoid molecular transporters. Proc. Natl Acad. Sci. 424(6944), 94–98 (2003).
USA 97(24), 13003–13008 (2000).
221 Sheehy AM, Gaddis NC, Malim MH. The antiretroviral 205 Wu C-H, Chen Y-P, Mou C-Y, Cheng R. Altering the enzyme APOBEC3G is degraded by the proteasome in Tat-derived peptide bioactivity landscape by changing the response to HIV-1 Vif. Nat. Med. 9(11), 1404–1407 (2003).
Arginine side chain length. Amino Acids 44(2), 473–480 (2013).
222 Stopak K, de Noronha C, Yonemoto W, Greene WC. HIV-1 Vif blocks the antiviral activity of APOBEC3G by impairing 206 Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. both its translation and intracellular stability. Mol. Cell. Nature 408(6810), 307–310 (2000).
12(3), 591–601 (2003).
207 Jeffrey PD, Gorina S, Pavletich NP. Crystal structure of the 223 Bogerd HP, Doehle BP, Wiegand HL, Cullen BR. A single tetramerization domain of the p53 tumor suppressor at 1.7 amino acid difference in the host APOBEC3G protein angstroms. Science 267(5203), 1498–1502 (1995).
controls the primate species specificity of HIV type 1 208 Clore GM, Omichinski JG, Sakaguchi K et al. High- virion infectivity factor. Proc. Natl Acad. Sci. USA 101(11), resolution structure of the oligomerization domain of p53 3770–3774 (2004).
by multidimensional NMR. Science 265(5170), 386–391 224 Mangeat B, Turelli P, Liao S, Trono D. A Single Amino Acid Determinant Governs the Species-specific Sensitivity 209 Longo F, Marchetti MA, Castagnoli L, Battaglia PA, of APOBEC3G to Vif Action. J. Biol. Chem. 279(15), Gigliani F. A novel approach to protein-protein interaction: complex formation between the P53 tumor suppressor and 225 He Z, Zhang W, Chen G, Xu R, Yu X-F. Characterization of the HIV Tat proteins. Biochem. Biophys. Res. Commun. Conserved Motifs in HIV-1 Vif Required for APOBEC3G 206(1), 326–334 (1995).
and APOBEC3F Interaction. J. Mol. Biol. 381(4), 1000– 210 Ariumi Y, Kaida A, Hatanaka M, Shimotohno K. Functional 1011 (2008).
cross-talk of HIV-1 Tat with p53 through Its C-terminal 226 Zhang L, Saadatmand J, Li X et al. Function analysis of domain. Biochem. Biophys. Res. Commun. 287(2), 556–561 sequences in human APOBEC3G involved in Vif-mediated degradation. Virology 370(1), 113–121 (2008).
211 Duan L, Ozaki I, Oakes JW, Taylor JP, Khalili K, Pomerantz Vif-A3G interaction as a basis for developing peptide-based
RJ. The tumor suppressor protein p53 strongly alters human immunodeficiency virus type 1 replication. J. Virol. 68(7), 4302–4313 (1994).
227 Mehle A, Wilson H, Zhang C et al. Identification of an APOBEC3G binding site in human immunodeficiency 212 Li CJ, Wang C, Friedman DJ, Pardee AB. Reciprocal virus type 1 Vif and inhibitors of Vif-APOBEC3G binding. modulations between p53 and Tat of human J. Virol. 81(23), 13235–13241 (2007).
future science group Review Chandra, Maes & Friedler 228 Reingewertz TH, Britan-Rosich E, Rotem-Bamberger S et al. 244 Debouck C. The HIV-1 protease as a therapeutic target for Mapping the Vif–A3G interaction using peptide arrays: a AIDS. AIDs Res. Hum. Retrovirus 8(2), 153–164 (1992).
basis for anti-HIV lead peptides. Bioorg. Med. Chem. 21(12), Strategies for anti-HIV-1 Protease drug development.
3523–3532 (2013).
245 Smith DB, Johnson KS. Single-step purification of 229 Schröfelbauer B, Chen D, Landau NR. A single amino acid polypeptides expressed in Escherichia coli as fusions with of APOBEC3G controls its species-specific interaction with glutathione S-transferase. Gene 67(1), 31–40 (1988).
virion infectivity factor (Vif). Proc. Natl Acad. Sci. USA 101(11), 3927–3932 (2004).
246 Sabbah EN, Druillennec S, Morellet N, Bouaziz S, Kroemer G, Roques BP. Interaction between the HIV-1 protein Vpr and 230 Donahue JP, Vetter ML, Mukhtar NA, D'Aquila RT. The the adenine nucleotide translocator. Chem. Biol. Drug. Des. HIV-1 Vif PPLP motif is necessary for human APOBEC3G 67(2), 145–154 (2006).
binding and degradation. Virology 377(1), 49–53 (2008).
247 Jacotot E, Ferri KF, El Hamel C et al. Control of 231 Dang Y, Wang X, York IA, Zheng Y-H. Identification of a Mitochondrial membrane permeabilization by adenine critical T(Q/D/E)x5ADx2(I/L) motif from primate lentivirus nucleotide translocator interacting with HIV-1 viral protein R Vif proteins that regulate APOBEC3G and APOBEC3F and Bcl-2. J. Expt. Med. 193(4), 509–520 (2001).
neutralizing activity. J. Virol. 84(17), 8561–8570 (2010).
248 Borgne-Sanchez A, Dupont S, Langonne A et al. Targeted 232 De Noronha CM, Sherman MP, Lin HW et al. Dynamic Vpr-derived peptides reach mitochondria to induce apoptosis disruptions in nuclear envelope architecture and integrity of [alpha]V[beta]3-expressing endothelial cells. Cell Death induced by HIV-1 Vpr. Science 294(5544), 1105–1108 Differ. 14(3), 422–435 (2006).
249 Kattenbeck B, Rohrhofer A, Niedrig M, Wolf H, Modrow 233 Levy DN, Fernandes LS, Williams W V, Weiner DB. S. Defined amino acids in the gag proteins of human Induction of cell differentiation by human immunodeficiency immunodeficiency virus type 1 are functionally active during virus 1 Vpr. Cell 72(4), 541–550 (1993).
virus assembly. Intervirology 39(1–2), 32–39 (1996).
234 Kogan M, Rappaport J. HIV-1 accessory protein Vpr: 250 Gallina A, Mantoan G, Rindi G, Milanesi G. Influence of MA relevance in the pathogenesis of HIV and potential for Internal sequences, but not of the myristylated N-terminus therapeutic intervention. Retrovirology 8(1), 25 (2011).
sequence, on the budding site of HIV-1 Gag protein. Biochem. 235 Emerman M. HIV-1, Vpr and the cell cycle. Curr. Biol. 6(9), Biophys. Res. Commun. 204(3), 1031–1038 (1994).
1096–1103 (1996).
251 Bukrinsky MI, Haggerty S, Dempsey MP et al. A nuclear 236 Ardon O, Zimmerman ES, Andersen JL, DeHart JL, localization signal within HIV-1 matrix protein that governs Blackett J, Planelles V. Induction of G2 arrest and binding infection of non-dividing cells. Nature 365(6447), 666–669 to cyclophilin A are independent phenotypes of human immunodeficiency virus type 1 Vpr. J. Virol. 80(8), 252 Dorfman T, Mammano F, Haseltine WA, Göttlinger HG. 3694–3700 (2006).
Role of the matrix protein in the virion association of the 237 Solbak S, Reksten T, Wray V et al. The intriguing cyclophilin human immunodeficiency virus type 1 envelope glycoprotein. A-HIV-1 Vpr interaction: prolyl cis/trans isomerisation J. Virol. 68(3), 1689–1696 (1994).
catalysis and specific binding. BMC Struct. Biol. 10(1), 31 253 Ota A, Tanaka-Taya K, Ueda S. Cross-reactivity of anti- HIV-1-p17-derivative peptide (P30–52) antibody to Env V3 238 Di Marzio P, Choe S, Ebright M, Knoblauch R, Landau NR. peptide. Hybridoma 18(2), 149–157 (1999).
Mutational analysis of cell cycle arrest, nuclear localization 254 Ota A, Liu X, Fujio H, Sakato Nobuo, Ueda S. Random and virion packaging of human immunodeficiency virus type expression of human immunodeficiency virus-1 (HIV-1) 1 Vpr. J. Virol. 69(12), 7909–7916 (1995).
pl7 (epitopes) on the surface of the HIV-1-infected cell. 239 Gaynor EM, Chen ISY. Analysis of apoptosis induced by Hybridoma 17(1), 73–75 (1998).
HIV-1 Vpr and examination of the possible role of the 255 Kohl NE, Emini EA, Schleif WA et al. Active human hHR23A protein. Expt. Cell Res. 267(2), 243–257 (2001).
immunodeficiency virus protease is required for viral 240 Franke EK, Yuan HEH, Luban J. Specific incorporation infectivity. Proc. Natl Acad. Sci. USA 85(13), 4686–4690 of cyclophilin A into HIV-1 virions. Nature 372(6504), 359–362 (1994).
256 Liu B, Dai R, Tian C-J, Dawson L, Gorelick R, Yu X-F. 241 Solbak SMØ, Wray V, Horvli O et al. The host-pathogen Interaction of the human immunodeficiency virus type 1 interaction of human cyclophilin A and HIV-1 Vpr requires nucleocapsid with actin. J. Virol. 73(4), 2901–2908 (1999).
specific N-terminal and novel C-terminal domains. BMC 257 Luciw PA. Human immunodeficiency viruses and their Struct. Biol. 11, 49 (2011).
replication. In: Field Virology (3rd Edition). Fields BN, Knipe 242 Luo Z, Butcher DJ, Murali R, Srinivasan A, Huang Z. DM, Howely PM, (Eds.), Philadelphia, PA, USA, Lippincott- Structural studies of synthetic peptide fragments derived Raven Publishers; 18 (1996).
from the HIV-1 Vpr protein. Biochem. Biophys. Res. Commun. 258 Misumi S, Kudo A, Azuma R, Tomonaga M, Furuishi 244(3), 732–736 (1998).
K, Shoji S. The p2gagPeptide, AEAMSQVTNTATIM, 243 BouHamdan M, Xue Y, Baudat Y et al. Diversity of HIV-1 processed from HIV-1 Pr55 Gag was found to be a suicide Vpr interactions involves usage of the WXXF Motif of host inhibitor of HIV-1 protease. Biochem. Biophys. Res. Commun. cell proteins. J. Biol. Chem. 273(14), 8009–8016 (1998).
241(2), 275–280 (1997).
Future Med. Chem. (2015) 7(8)
future science group Interactions of HIV-1 proteins as targets for developing anti-HIV-1 peptides Review 259 Kotler M, Simm M, Zhao YS et al. Human immunodeficiency 272 Kilby JM, Hopkins S, Venetta TM et al. Potent suppression virus type 1 (HIV-1) protein Vif inhibits the activity of HIV-1 of HIV-1 replication in humans by T-20, a peptide inhibitor protease in bacteria and in vitro. J. Virol. 71(8), 5774–5781 of gp41-mediated virus entry. Nat. Med. 4(11), 1302–1307 260 Karageorgos L, Li P, Burrell C. Characterization of HIV 273 White CJ & Yudin AK. Contemporary strategies for peptide replication complexes early after cell-to-cell infection. AIDS macrocyclization. Nat. Chem. 3, 509–524 (2011).
Res. Hum. Retroviruses 9(9), 817–823 (1993).
274 Kazmierski WM, Kenakin TP, Gudmundsson KS. Peptide, 261 Potash MJ, Bentsman G, Muir T, Krachmarov C, Sova P, peptidomimetic and small-molecule drug discovery targeting Volsky DJ. Peptide inhibitors of HIV-1 protease and viral HIV-1 host-cell attachment and entry through gp120, gp41, infection of peripheral blood lymphocytes based on HIV-1 CCR5 and CXCR4†. Chem. Biol. Drug. Des. 67(1), 13–26 Vif. Proc. Natl Acad. Sci. USA 95(23), 13865–13868 (1998).
262 Baraz L, Hutoran M, Blumenzweig I et al. Human 275 Cai L, Jiang S. Development of peptide and small-molecule immunodeficiency virus type 1 Vif binds the viral protease by HIV-1 fusion inhibitors that target gp41. ChemMedChem interaction with its N-terminal region. J. Gen. Virol. 83(9), 5(11), 1813–1824 (2010).
2225–2230 (2002).
276 Brunton LL, Lazo JS, Parker K. Goodman and Gilmans´s 263 Friedler A, Blumenzweig I, Baraz L, Steinitz M, Kotler M, The Pharmacological Basis of Therapeutics (11th Edition). Gilon C. Peptides derived from HIV-1 vif: a non-substrate McGraw-Hill, USA (2006).
based novel type of HIV-1 protease inhibitors. J. Mol. Biol. 277 Flexner C. HIV drug development: the next 25 years. Nat. 287(1), 93–101 (1999).
Rev. Drug Discov. 6(12), 959–966 (2007).
264 Hutoran M, Britan E, Baraz L, Blumenzweig I, Steinitz M, 278 Wlodawer A. Rational approach to AIDS drug design Kotler M. Abrogation of Vif function by peptide derived from through structural biology. Annu. Rev. Med. 53(1), 595–614 the N-terminal region of the human immunodeficiency virus type 1 (HIV-1) protease. Virology 330(1), 261–270 (2004).
279 Liu J, Bartesaghi A, Borgnia MJ, Sapiro G, Subramaniam 265 Gallay P, Swingler S, Song J, Bushman F, Trono D. HIV S. Molecular architecture of native HIV-1 gp120 trimers. nuclear import is governed by the phosphotyrosine-mediated Nature 455(7209), 109–113 (2008).
binding of matrix to the core domain of integrase. Cell 83(4), 569–576 (1995).
280 Dwyer JJ, Wilson KL, Martin K et al. Design of an engineered N-terminal HIV-1 gp41 trimer with enhanced 266 Oz Gleenberg I, Avidan O, Goldgur Y, Herschhorn A, Hizi stability and potency. Protein Sci. 17(4), 633–643 (2008).
A. Peptides Derived from the Reverse transcriptase of human immunodeficiency virus type 1 as novel inhibitors of the viral 281 Das K, Martinez SE, Bauman JD, Arnold E. HIV-1 reverse transcriptase complex with DNA and nevirapine reveals integrase. J. Biol. Chem. 280(23), 21987–21996 (2005).
non-nucleoside inhibition mechanism. Nat. Struct. Mol. Biol. 267 Bischerour J, Tauc P, Leh H, Rocquigny H de, Roques 19(2), 253–259 (2012).
B, Mouscadet J. The (52–96) C-terminal domain of Vpr stimulates HIV-1 IN-mediated homologous strand transfer 282 Kitamura S, Ode H, Nakashima M et al. The APOBEC3C crystal structure and the interface for HIV-1 Vif binding. of mini-viral DNA. Nucleic Acids Res. 31(10), 2694–2702 (2003).
Nat. Struct. Mol. Biol. 19(10), 1005–1010 (2012).
283 Shandilya SMD, Nalam MNL, Nalivaika EA et al. Crystal 268 Catherine S. Adamson EOF. Anti-HIV-1 therapeutics: from Structure of the APOBEC3G Catalytic Domain Reveals FDA-approved Drugs to Hypothetical Future Targets. Mol. Potential Oligomerization Interfaces. Interv. 9(2), 70–74 (2009).
Structure 18(1), 28–38 269 Pang W, Tam S-C, Zheng Y-T. Current Peptide HIV Type-1 Fusion Inhibitors. Antivir. Chem. Chemother. 20(1), 1–18 284 Wielens J, Headey SJ, Jeevarajah D et al. Crystal structure of the HIV-1 integrase core domain in complex with sucrose reveals details of an allosteric inhibitory binding site. FEBS 270 Park M, Wetzler M, Jardetzky TS, Barron AE. A readily Lett. 584(8), 1455–1462 (2015).
applicable strategy to convert peptides to peptoid-based therapeutics. PLoS ONE 8(3), e58874 (2013).
285 Im YJ, Kuo L, Ren X et al. Crystallographic and functional analysis of the ESCRT-I /HIV-1 Gag PTAP interaction. 271 Henchey LK, Jochim AL, Arora PS. Contemporary strategies Structure 18(11), 1536–1547 (2015).
for the stabilization of peptides in the α-helical conformation. Curr. Opin. Chem. Biol. 12(6), 692–697 (2008).
future science group

Source: http://chem.ch.huji.ac.il/~assaf/papers/2015_Chandra_et_al_FMC.pdf

Eurogames_konferenz_d_neu

Europäische Konferenz über Homosexualität und Behinderung im Sport Behinderte bei den EuroGames 2004 Vision: Brücken bauen Menschen mit Behinderungen sind ebenso wie Schwule, Les- die im Rahmen der EuroGames in München stattfand, war es, die ben, Bi- und Transsexuel e in der Gesel schaft mit spezifi schen Themenbereiche Homosexualität, Behinderung und Sport aufzu-

unl.edu

Race and Medicine Genetic studies of population differences, although controversial, promise David Goldstein of University College in clues to disease as well as new drug targets, scientists believe London agrees: "If you say on average the difference between West Africans and Eu- Mention race and medicine in a group of racial identity biologically irrelevant. But ropeans is slight, that does not rule out a