ArticlePDF AvailableLiterature Review

Proteolytic Events of HIV-1 Replication as Targets for Therapeutic Intervention

Authors:

Abstract and Figures

Acquired immunodeficiency syndrome (AIDS) is a worldwide epidemic caused by infection with HIV, a human retrovirus. Proteolysis occurs at many points of the retroviral life-cycle, and these events can be considered as targets for chemotherapy. The most well-known proteolytic action in the retroviral life-cycle is the processing of the Gag and Gag-Pro-Pol polyproteins with the virally encoded protease at the late phase of viral infection. Protease inhibitors, together with reverse transcriptase inhibitors, are important components of the drug combinations currently used to treat HIV patients. The current combination therapy substantially reduced morbidity and mortality in HIV-infected patients. However, these drugs do not allow viral eradication, therefore their long-term use is required, allowing the development of resistance in a large portion of patients. Furthermore, several adverse metabolic side effects have been observed associated with the therapy. Thus, new approaches are required to eradicate HIV infection, which may include targeting of the potential early-phase function of the viral protease, and other crucial proteolytic events of the viral replication, such as the ubiquitin-dependent proteolytic degradation of the unfolded viral proteins as well as the inhibition of envelope protein processing.
Content may be subject to copyright.
Current Pharmaceutical Design, 2003, 9, 1803-1815 1803
1381-6128/03 $41.00+.00 © 2003 Bentham Science Publishers Ltd.
Proteolytic Events of HIV-1 Replication as Targets for Therapeutic
Intervention
J. Tözsér1,* and S. Oroszlan2
1Department of Biochemistry and Molecular Biology, Faculty of Medicine, Debrecen University, H-4012 Debrecen,
Hungary and 2HIV Drug Resistance Program, National Cancer Institute at Frederick, Frederick, MD 21701, USA
Abstract: Acquired immunodeficiency syndrome (AIDS) is a worldwide epidemic caused by infection with HIV, a
human retrovirus. Proteolysis occurs at many points of the retroviral life-cycle, and these events can be considered as
targets for chemotherapy. The most well-known proteolytic action in the retroviral life-cycle is the processing of the Gag
and Gag-Pro-Pol polyproteins with the virally encoded protease at the late phase of viral infection. Protease inhibitors,
together with reverse transcriptase inhibitors, are important components of the drug combinations currently used to treat
HIV patients. The current combination therapy substantially reduced morbidity and mortality in HIV-infected patients.
However, these drugs do not allow viral eradication, therefore their long-term use is required, allowing the development
of resistance in a large portion of patients. Furthermore, several adverse metabolic side effects have been observed
associated with the therapy. Thus, new approaches are required to eradicate HIV infection, which may include targeting of
the potential early-phase function of the viral protease, and other crucial proteolytic events of the viral replication, such as
the ubiquitin-dependent proteolytic degradation of the unfolded viral proteins as well as the inhibition of envelope protein
processing.
Key Words: HIV-1, life-cycle, proteolysis, viral protease, envelope protein, ubiquitin, proteasomal degradation.
1. INTRODUCTION
Several anti-retroviral drugs are available for clinical use
to treat AIDS patients, and the most commonly used
treatment involves the combination of inhibitors of two
replication enzymes of human immunodeficiency virus type
1 (HIV-1), reverse transcriptase (RT) and protease (PR). The
use of these drugs in combination (a treatment termed highly
active antiretroviral therapy, HAART) substantially reduces
morbidity and mortality in HIV-infected patients (Reviewed
in [1]). Unfortunately this combination therapy is expensive,
therefore it is not readily available for patients in developing
countries, and it does not allow viral eradication. Due to the
lack of virus elimination, long-term use of these drugs is
required, allowing the virus to mutate into drug-resistant
forms in a large portion of patients. The existence of organ
sanctuaries (e.g., brain, testis) and long-lived infected cells
carrying proviral DNA contribute to the limits of eradicating
the virus (for a review see [2]). Most of the problems
associated with the HIV therapy are the consequence of the
necessarily long-term use of the drugs. Besides the
appearance of resistance several metabolic side effects exist,
including hypersensitivity, lipodystrophy syndrome, insulin
resistance and cardiovascular diseases (for reviews see [3-
5]).
Therefore it is essential to find drugs targeting the same
enzymes but with improved characteristics, or drugs targeted
to alternative steps of the viral replication cycle. Most of the
steps of the retroviral life-cycle could be targeted, and being
*Address correspondence to this author at the Debrecen University, H-4012
Debrecen, P. O. Box 6, Hungary. E-mail: tozser@indi.biochem.dote.hu
explored to provide novel anti-retroviral drugs. The aim of
this review is to summarize the essential proteolytic events in
viral replication, as well as the current status and future
prospects of the use of protease inhibitors.
2. OVERVIEW OF THE RETROVIRAL REPLICA-
TION CYCLE
To help the understanding of the proteolytic target points
in HIV infection, a short description of the retroviral life-
cycle is presented below (Fig. 1). In the early phase of the
cycle, the virus enters the host cell by membrane fusion or
receptor-mediated endocytosis (step 1). Then reverse
transcription occurs in the entering capsid structure, in which
the plus-strand RNA genome is converted to a double-
stranded DNA by RT (step 2). The error-prone nature of the
RT typically results in one mutation/genome in this step;
therefore, the virus is considered as a quasi-species. The high
mutation rate, together with the high recombination capacity
of the virus, allows the development of resistance towards
any kind of inhibitors targeting viral enzymes or proteins.
Following the reverse transcription, a preintegration complex
(PIC) is formed, composed of the genomic DNA and some
of the protein components of the entering capsid. The PIC
enters the nucleus (step 3). This step is typically a passive
process in case of most retroviruses, since they can infect
only dividing cells, in which the nuclear envelope is
disassembled; however, in the case of HIV-1 and other
retroviruses belonging to the lentiviral subfamily, an active
transport of the PIC allows the infection of nondividing cells.
The integration of the viral DNA into the host genome (step
4) is mediated by the integrase (IN), which is an essential
part of the PIC. The late-phase of the retroviral life-cycle
1804 Current Pharmaceutical Design, 2003, Vol. 9, No. 22 Tözsér and Oroszlan
starts with the transcription of viral DNA into various RNA
forms by the host RNA polymerase II (step 5). A significant
fraction of newly synthesized mRNA reaches the cytoplasm
unspliced, to serve as template for the Gag and Gag-Pro-Pol
polyproteins (step 6), and to be packaged into virions as
genomic RNA. A singly spliced RNA serves as a template
for the envelope protein (Env) synthesis in the endoplasmic
reticulum and Golgi. The Env protein is processed by a
cellular enzyme into a surface protein (SU) and a transmem-
brane protein (TM), and exported to the plasma membrane,
to the site of assembly. Various singly and multiply spliced
RNAs serve as templates for the synthesis of accessory
proteins, such as Tat, Rev, Vif, Nef, and Vpr. The Gag and
Gag-Pro-Pol polyproteins assemble at the membrane where
the Env proteins are concentrated (step 7); then the
assembled "immature" particle buds from the membrane
(step 8), followed by "maturation" to a morphologically
distinct form with a condensed core (step 9).
3. PROTEOLYTIC EVENTS IN THE LATE-PHASE
OF RETROVIRAL REPLICATION
3.1. Function and Properties of the Virally Encoded
Protease
3.1.1. Polyprotein Processing by the Retroviral Protease
In the last stage of the late phase of viral replication, the
Gag and Gag-Pro-Pol polyproteins are assembled together
with the envelope proteins and the viral genomic RNA at the
plasma membrane of the infected cell, and budding yields
“immature” virions with a “doughnut-shaped” capsid
structure (Fig. 1). The PR cleaves the viral polyproteins at a
limited number of sites (Fig. 2A), producing a condensed,
cone-shaped core of “mature” infectious HIV-1 (Fig. 1). This
function of the PR is essential for virus replication. Recent
reports suggest that redox regulation may be involved in PR
activation and subsequent “maturation” [6-8]. As shown in
Fig. 2A, the PR is coded on the pro gene and is part of the
Gag-Pro-Pol polyprotein synthesized by ribosomal
frameshifting (Reviewed in [9]). The relatively low
frequency of frameshifting assures that the amount of
replication enzymes in the virions is only about 5-10 % of
those of the structural proteins encoded by the gag gene. The
cleavage sites where the polyproteins are processed are given
in Fig. 2B. For comparison, cleavage sites verified within
some viral proteins as well as cleavage sites in encapsidated
cellular proteins, are also shown here.
3.1.2. Properties and Specificity of the Retroviral Protease
The structure of the HIV-1 PR was determined more than
a decade ago, and currently it is the most represented
structure in the protein structural databanks (for reviews see
[10,11]). The HIV-1 PR is a homodimeric aspartic protease
of 99-residue-long subunits, and in the absence of a ligand, it
is almost perfectly symmetrical. It has two flap regions that
are flexible in the absence of the ligand, but they are closed
Fig. (1). Replication cycle of HIV-1. The cycle starts by the interaction of the envelope protein with the receptor and coreceptor on the
surface of susceptible cell, leading to fusion and entry of the cone-shaped core, or alternatively, the virus may enter into the cell by receptor-
mediated endocytosis (step 1). Reverse transcription (step 2) generates a double-stranded DNA copy of the RNA genome. The provirus is
transported into the nucleus (step 3) and integrated (step 4) into chromosomal DNA. In the late phase of the cycle, the viral DNA is
transcribed by the cellular RNA polymerase II (step 5). Unspliced RNA is used as template for the synthesis of structural proteins and
replication enzymes (step 6), while the Env precursor protein is synthesized from spliced RNA in the endoplasmic reticulum and Golgi.
Virion proteins and progeny RNA assemble at the plasma membrane (step 7), and progeny “immature” virus is released by a process of
budding (step 8) followed by PR-mediated “maturation” (step 9) into infectious virus.
Proteolytic Events of HIV-1 Replication Current Pharmaceutical Design, 2003, Vol. 9, No. 22 1805
Fig. (2). Schematic representation of the HIV-1 genome (A) and sequences of HIV protease cleavage sites (B).
The location of open reading frames is indicated. The RNA encapsidation signal (ψ) is shown near the 5' end. The proteins encoded by the
pro and pol genes are synthesized by frameshifting mechanism from genome-sized RNA template to yield Gag-Pro-Pol polyproteins. Gag and
Gag-Pro-Pol polyproteins are processed at the last step of viral life-cycle by the viral PR at the sites indicated by arrows above the boxes. Env
is also synthesized as a precursor (SU + TM) and processed at the indicated site by a cellular protease.
The Gag polyproteins are processed into matrix (MA), capsid (CA), nucleocapsid (NC), and three smaller fragments and proteins designated
according to their molecular weight (e.g. p1, a one kDa peptide), while cleavage of Gag-Pro-Pol polyproteins also yields transframe protein
(TF), protease (PR), reverse transcriptase (RT) which is active as p66/p51 having RNase H (RH) domain in the larger protein, and integrase
(IN). Cleavages within these proteins which may occur in the early-phase of viral life-cycle are indicated by arrows below the boxes.
Classification of cleavage sites was done based on the sequence around the site of cleavage.
1806 Current Pharmaceutical Design, 2003, Vol. 9, No. 22 Tözsér and Oroszlan
down on the bound inhibitors as well as substrates (Fig. 3).
The activity of the PR is mediated by the active site aspartate
residues, which are held in a rigid position by a network of
hydrogen bonds called the “fireman’s grip”. The enzyme has
an acidic pH optimum, but its actual value depends on the
substrate [12].
Fig. (3). Ribbon diagram of the active protease dimer with a bound
ligand. The ligand is shown in ball-and-stick representation.
It was already recognized in the early stage of HIV PR
studies that understanding of its specificity is crucial for the
design of efficient inhibitors. Numerous studies have been
done on the specificity of the HIV-1 PR (For reviews see
[13-16]). It is a special feature of the PR, that it is not
possible to give a consensus substrate sequence, even though
the enzyme is fairly specific. Retroviral protease cleavage
sites are currently classified into two groups [17-19]. Type 1
c le ava ge s ite s have a roma tic re sidue a nd Pro, and type 2 s ite s
have hydrophobic res idues (e xc luding Pro) a t the s ite of
c le ava ge (Fig. 2B). The P2 a nd P2’ pos itions (nota tion is
a cc ording to [20]) a re als o critica l in determining the
s pe cific ity [18,19]. In type 1 clea vage s ites of primate
lentivirus e s, like H IV-1, there is a pre ferenc e for A sn a t P2
a nd be ta -branche d hydrophobic res idue (V a l or Ile ) a t P2’,
w hile in type 2 clea vage s ites the P2 pos ition is typic a lly beta
bra nc hed a nd the P2’ re sidue is G lu or G ln (Fig. 2B ). The type
1 c le a va ge site is ve ry importa nt for se veral re as ons . No other
protease, except pepsin, is known to act at the imino side of a
Pro residue. Indeed, the uniqueness of Pro in that position
was recognized even before the discovery of the PR [21] and
it was suggested early that inhibitors based on the this type
of cleavage site should be specific against the retroviral PRs
[22]. Proline residues, especially after Tyr or Phe in the
sequence (as the case in type 1 cleavage sites) have a
relatively high probability of forming the cis isomer rather
than the trans isomer of the preceding peptide bond [23] and
molecular modeling studies indicated that a trans rather than
the cis isomer provided a better fit into the substrate-binding
pocket [19]. Based on these findings we inferred that
isomerization at the proline of retroviral cleavage sites may
be involved in the triggering of maturation events [19].
Subsequently the conformational selectivity of the HIV-1 PR
towards the trans isomer of the cleaved peptide bond was
demonstrated by NMR and kinetic studies [24,25].
A schematic diagram of the substrate binding site of
HIV-1 PR with modeled interaction of the bound residues of
the MACA cleavage site is given in Fig. 4A. The substrate
binds to the enzyme in an extended beta conformation, and it
is anchored by several hydrogen bonds. Substrate-based
inhibitors bind in a very similar manner. The enzyme
recognizes at least seven substrate residues, from P4 to P3’
(Fig. 4A). Based on detailed specificity studies as well as
HIV-1 PR-inhibitor crystal structures, there appear to be a
very strong sequence context dependence of the specificity
of PR. In other terms, the interactions between the ligand
side chains and respective binding pockets are not additive;
various types of ligand side chain interactions complicate
them [26]. The internal substrate side chain interactions that
affect the preference at a given subsite can be classified into
three types (Fig. 4B). In a cis interaction the substrate side
chains positioned at the same side of the substrate main
chain influence the positions of each other; for example, a
beta-branched side chain at P2 diminishes the place available
for the P4 side chain. In a trans interaction sequential
residues being at the opposite site of the substrate main chain
influence each other’s positions. For example, a bulky side
chain (like Tyr) at P1’ reduces the space available for the P1
and P2’ side chains by pushing the peptide backbone of the
substrate toward those sites. In a coupled interaction a substi-
tution of a side chain causes a sequential rearrangement of
other substrate side chains and could therefore influence the
preference for a distant site. Such interactions can cause the
effect of the P3’ side chain on P2 [26]. The strong s equence
context-depe ndenc e should be taken into ac count in the de sign
of protea se inhibitors : a mutation in a subs tra te binding subs ite
of the PR indirec tly c ould influence the s pe cific ity of the othe r
binding s ite s.
As discussed above, HIV-1 PR cleaves the Gag and Gag-
Pro-Pol proteins at various positions during viral maturation.
Although only one, a typical type 2 viral HIV-1 PR
maturation cleavage site, the CAp2 site, contains charged
residue at the P2-P2’ region, a P2' glutamate, this residue is
highly conserved in HIV-1 [27]. The cleavage at this site is
accelerated by lower pH, and this was suggested to play a
regulatory role in the viral protein processing [28].
Crystallographic structures of HIV proteases have been
reported with inhibitors having P2’ Glu residues [29,30]. In
these structures the P2’ Glu showed two weak hydrogen
bond interactions with the amides of Asp 29 and Asp 30 of
the enzyme. The proximity of the Glu side chain to the side
chain of Asp 30 suggested that they share a proton [30]. The
presence of glutamate at the P2' position of various peptide
substrates was found to be responsible for the pH effect [31].
Interestingly, the cleavage site within the N-terminal domain
of p24 contains Glu residue at both the P2 and P2' positions
(Fig. 2B). The presence of two glutamates in this site
enhanced its sensitivity to pH [32]. Unlike the P2 and P2'
glutamates, Glu at the P3 and P3’ positions (as in the
cleavage site in the C-terminal domain of CA) did not result
in such an effect [32, our unpublished results]. It is of
interest to note, that unlike in the Gag and Gag-Pro-Pol
cleavage sites, cellular protein cleavage sites frequently
Proteolytic Events of HIV-1 Replication Current Pharmaceutical Design, 2003, Vol. 9, No. 22 1807
contain charged residues, especially Glu at P2’ [13]. The
reason for the markedly different subsite preference in viral
and cellular proteins of the HIV-1 PR is not known.
3.1.3. HIV Protease Inhibitors and Development of
Resistance Against Them
The names and structures of HIV PR inhibitors currently
used in therapy are shown in Fig. 5. A common feature of
these compounds is that they are peptidomimetics; they
mimic the substrates of the PR. The enzyme-inhibitor
interactions, similar to the enzyme-substrate interactions, are
primarily hydrophobic ones. Typically these inhibitors
contain a phenyl residue at the P1 position. Another common
feature of these inhibitors is that they contain a nonhydro-
lyzable transition state mimic, like a hydoxyethylamine
group at the site corresponding to the cleavable bond in the
Fig. (4). Schematic representation of the substrate binding subsites of the HIV-1 protease (A) and type of internal substrate side chain
interactions (B).
The relative size of each subsite is indicated approximately by the area enclosed by the curved line around each substrate side chain.
According to the nomenclature, residues of the ligand (substrate or inhibitor) interacting with the protease are designated as P1, P2, P3 etc.
from the scissile bond towards the N-terminus of the substrate, and P1’, P2’, etc. towards the C-terminus. The respective substrate binding
subsites are designated as S1, S2, etc. The relative location of residues forming the subsites (see Fig. 6) is also shown. Some of the mutations
occurring in resistance are indicated as wild type residue/residue found in resistant viruses. Amino acid residues of the second subunit of the
protease dimer are indicated by a prime. The substituted residues are represented by open, dashed-lined objects in panel B, and the induced
conformation of the other side chains is represented by open, thick-lined objects.
1808 Current Pharmaceutical Design, 2003, Vol. 9, No. 22 Tözsér and Oroszlan
substrates. Saquinavir was the first approved HIV-1 PR drug.
It is based on a type 1 cleavage site, in which the Pro was
replaced by a saturated isoquinoline ring. Indinavir and
nelfinavir also mimic the type 1 cleavage site; ritonavir was
developed from a symmetric molecule, while amprenavir is a
sulfonamide compound. Lopinavir is the first second-
generation PR inhibitor. Its core is identical to that of
ritonavir, and it was designed to diminish the interactions of
the inhibitor with Val 82, a residue that is frequently mutated
in drug-resistant strains of the virus [33]. It is used in
combination with a smaller amount of ritonavir.
Resistance develops against PR inhibitors. In most of the
cases, mutations occur in the PR gene (Fig. 6A). While most
of the natural sequence variations (that is residues found in
different HIV-1 clones in the absence of PR inhibitor
treatment) are outside of the substrate binding sites, several
of the mutations conferring resistance involve residues of the
substrate binding subsites; therefore, they are expected to
alter the specificity and catalytic power of the enzyme.
Recent studies showed, that in patients receiving HIV PR
inhibitors, not only is the PR mutated, but mutations were
also observed at the NCp1 and p1p6 Gag cleavage sites
(Fig. 6B). These mutations were first described in in vitro
studies, but later they were also found in patients undergoing
indinavir therapy [34-36]. These mutations occurred together
with mutations in the PR gene [36]. Evolution of PR
cleavage sites other than NCp1 and p1p6 in the internal
(P2-P2') positions is limited, and mutations are rarely
observed even upon drug treatment [37]. Cleavage at these
sites appears to be a rate-limiting step in polyprotein
processing [34,38]. Peptides representing these sites have the
Fig. (5). Name and structure of HIV-1 protease inhibitors currently used in antiretroviral therapy.
N
H
NN
H
O
O
O
H2N
N
OH
O NH
Saquinavir
Invirase
Ro 31-8959
N
N
NH
N
OH
O
OH
NHO
Indinavir
Crixivan
MK-639
L735,524
NNH
NN
H
S
O
OH
NO
OH
O
S
N
Ritonavir
Norvir
ABT-538
N
HN
OH
O
OH
SNHO
Nelfinavir
Viracept
Ag-1343
O
O N
H
O
NSOO
NH2
OH
Amprenavir
Aguerase
VX-478
HN N
H
N
O
O OH
N
H
O
O
Lopinavir
Aluviran
ABT-378
Proteolytic Events of HIV-1 Replication Current Pharmaceutical Design, 2003, Vol. 9, No. 22 1809
lowest specificity constants (kcat/Km) among all HIV-1
cleavage sites [39,40]. Furthermore, there is a significant
sequence polymorphism at these sites (Fig. 6B), which also
may have an impact on virion infectivity [27]. Recently we
have studied the hydrolysis of oligopeptides representing
these cleavage sites with representative mutations found as
natural variations or that arise as resistant mutations [41].
Wild-type and five drug-resistant PRs with mutations within
or outside the substrate binding site were tested. While the
natural variations showed either increased or decreased
susceptibility of peptides toward the PR, the resistant muta-
tions always had a beneficial effect on catalytic efficiency
[41]. Comparison of the specificity changes obtained for the
various substrates suggested that the maximization of the van
der Waals contacts between substrate and PR is the major
determinant of specificity: the same effect is crucial for
inhibitor potency. Resistant mutants of HIV-1 PR must
possess sufficient proteolytic activity (kcat/Km) to support
viral replication by correctly cleaving Gag and Gag-Pro-Pol
precursors. The order of Gag cleavage is also important for
infectivity [42]. Unlike our findings with the MACA and
CANC cleavage sites [19,31], the natural NCp1 and
p1p6 sites do not appear to be optimized for rapid
hydrolysis. Therefore, it is possible to increase the cleavage
rate by mutation at these sites when the PR activity is
diminished due to the accumulation of PR mutations. Hence,
mutation of these rate-limiting cleavage sites can partly
compensate for the reduced catalytic activity of drug-
resistant mutant HIV-1 proteases.
3.2. Processing of the Envelope Protein
Viral surface glycoproteins are involved in both binding
and penetration of the virus into the target cells (reviewed by
[43,44]). Like the Gag and Gag-Pro-Pol, these glycoproteins
are also synthesized as precursors, and sorted through the
secretory pathway of the infected cells, where they are
cleaved at a highly conserved basic consensus sequence
(Arg-Xaa-Lys/Arg-Arg--Xaa) (for a review see [45]). This
cleavage leads to the formation of the mature surface (SU)
and transmembrane (TM) glycoproteins that remain
Fig. (6). Sequence variation of HIV-1 protease (A) and the Gag region around p1 (B). The protease sequence of the HXB2 clone is given, the
conserved regions characteristic of retroviral proteases are indicated by bold, larger letters. Residues of the protease, which are involved in
ligand binding, are underlined. Residues occurring as natural variations are indicated by lower case letters in both sequences, while those
which have also been found as mutations occurring in drug resistance are in bold. Residues occurring only in drug resistant mutants are
indicated by capital bold letters.
1810 Current Pharmaceutical Design, 2003, Vol. 9, No. 22 Tözsér and Oroszlan
associated by noncovalent interactions. The SU recognizes
the host-cell receptors, and then the envelope complex
undergoes a conformational change that leads to exposure of
the N-terminal fusion peptide of the TM, which then
promotes virus-cell fusion in the classical infection route.
The cleavage and maturation of the envelope glycoproteins
are required for infectivity [46]. This processing is
performed by enzymes belonging to the Ca2+-dependent
prohormone convertase (PC) family, including furin, PC7,
PACE4 (paired amino acids-cleaving enzyme 4), and PC5
(reviewed in [45]). Calcium-independent enzymes may also
be involved in this process [47,48]. So far two types of
inhibitors have been developed to inhibit the Env cleavage.
An oligopeptide-chloroalkylketone derivative [49] and an
inhibitor derived from serine protease inhibitors (serpins) by
mutagenesis [50]. Due to the high variability of HIV and the
inevitable appearance of resistant mutants against the drugs
targeting the viral proteins, it may be beneficial to utilize
drugs targeting the cellular proteins and enzymes involved in
the life-cycle.
An additional cleavage within the V3 loop of the SU was
reported [51], and the enzymes were later identified as T-cell
membrane-associated serine protease, tryptase TL2 [52], and
urokinase-type plasminogen activator in case of
macrophages [53]. The significance of the V3 cleavage in
infection is not completely understood, it is proposed to
enhance the fusion process [53].
3.3. The Role of Ubiquitin and Ubiquitin-Dependent
Proteasomal Degradation in Assembly
Ubiquitin is a small, highly conserved protein that has
several functions in the eukaryotic cells. The classical role of
ubiquitination is to target the tagged proteins for degradation
by a multisubunit ATP-dependent protease termed the
proteasome. Ubiquitination is also emerging as a signal for
trafficking inside the cell. The first report that retroviral
particles contain ubiquitin was in 1988 when it was detected
in HIV as previously seen with murine leukemia virus
(MuLV) particles [54]. Free, and late assembly domain-
conjugated (p6-conjugated in HIV-1) ubiquitin was also
found in simian immunodeficiency virus (SIV), MuLV and
EIAV [55,56], while avian leukosis virus contains only free
ubiquitin [57]. Although these late assembly domain-
containing proteins appear to be monoubiquitinated, which is
not the typical protein tagging for degradation, it has been
shown, that the use of proteasome inhibitors, which deplete
the free ubiquitin pool by entrapping ubiquitins in
conjugated forms, inhibit HIV-1 budding and hence viral
replication [58]. On the other hand, EIAV budding was not
inhibited by proteasome inhibitors [56]. Interestingly, the
proteasome inhibitors also reduced Gag maturation in HIV-
1, thus influencing the processing by PR [58]. One possible
mechanism is the buildup of defective Gag translation
products that otherwise would be rapidly degraded by the
proteasome [58]. A large portion (even one-third) of newly
synthesized cellular proteins cannot fold properly, and are
degraded by the proteasome, and the same could be valid for
the viral proteins [59]. Kisselev and Goldberg [60] reviewed
several type of inhibitors developed against the proteasomes.
Ritonavir and saquinavir, inhibitors used against HIV-1 PR
also can inhibit proteasomes [61,62]. However, since the
proteasomal degradation has a protective function in the
early-phase, use of proteasomal inhibitors may have dubious
effects on HIV replication.
4. PROTEOLYTIC EVENTS IN THE EARLY PHASE
OF RETROVIRAL REPLICATION
Unlike the late-phase of retroviral replication, the early
phase is much less understood, mostly due to the difficulties
in finding proper techniques and detection systems, since
only a small fraction of the entering virions is infectious. It
has been a paradigm in retrovirus research that unlike most
enveloped viruses including some retroviruses using the
receptor-mediated endocytotic route, HIV enters the cell
utilizing a pH-independent direct fusion of the virion and the
cell membrane (Reviewed in [44]). However, the distinction
between entry of pH-independent and pH-dependent viruses
is not as simple as it was originally thought. Several cases of
pH-independent viruses that appear to require endocytosis
for productive infection have now emerged [63].
Amphotropic and ecotropic murine leukemia viruses also use
the endocytic route, but the infection is pH independent [64].
Although HIV was shown to be able to enter the cells by
clathrin-coated vesicles [65] and a large fraction of HIV-1
can be taken up by the cell via endocytosis [66,67], it was
shown that this pathway usually does not result in productive
infection, presumably due to the inability of the virion to
escape from the endosome [66,67]. However, it has been
recently demonstrated that endocytotic entry of HIV-1 into
cells can lead to viral integration and gene expression [68].
The use of direct fusion and the endocytotic route depends
on the type of virus producing cells, target cells and viral
isolates [68]. Primary macrophages mostly take up HIV-1 by
macropinocytosis, a receptor-independent process [69].
Following uptake, a large part of the macropinocytosed
virions was degraded, likely because of
endosomal/lysosomal fusions [69] but productive infection
also occurred. This can be explained by the envelope-
mediated fusion of some HIV-1 virions with the vesicular
membrane. The infection route used by the virion may have
a profound effect on the proteolytic events and proteolytic
enzymes involved in the early phase of viral replication.
4.1. Potential Early Phase Proteolytic Events Mediated
by the Viral Protease
While the late-phase function of the virally coded
protease is well established, its function in the early phase is
controversial and much debated. It was first demonstrated for
equine infectious anemia virus (EIAV) [70] and later for
HIV [71] that the PR is part of the core structure, which
enters the infected cells. In both the receptor-mediated
endocytotic and macropinocytotic routes, the incoming core
is surrounded by an acidic environment, which has an
optimal pH for the retroviral protease.
4.1.1. Cleavage of Proteins of the Host Cell by the Viral
Protease
It has been observed, that the HIV PR is capable of
cleaving cytoskeletal and sarcomeric proteins including
vimentin, desmin, actin, myosin and tropomyosin [72,73]. It
has been proposed that the processing of vimentin is
Proteolytic Events of HIV-1 Replication Current Pharmaceutical Design, 2003, Vol. 9, No. 22 1811
important in the early phase of viral infection [74].
Interestingly, while the actin cleavage was regulated by
polymerization/depolymerization [73], the vimentin cleavage
can be regulated by phosphorylation: only the
dephosphorylated form of the protein is a substrate of the PR
[75]. The journey of the core towards the nucleus requires
interaction with the actin microfilaments [76]; therefore
proteolysis of the components of the actin microfilament
may be an important step in regulation of this process. The
cleavage of other cellular proteins, like the transcription
factor NF-κB and eukaryotic initiation factor eIF4GI, may
rather be important in the late phase of viral replication, by
affecting gene transcription and translation, as well as the
cytotoxicity of the virus [77,79].
4.1.2. Cleavage of Viral and Virion-Packaged Cellular
Proteins by the Viral Protease
Besides the mature PR, the core of HIV-1 contains
several viral proteins, including capsid (CA), nucleocapsid
(NC), reverse transcriptase (RT), RNase H, integrase (IN),
Vpr and Nef [71]. Cellular proteins were also detected in
virions including the cellular peptidyl prolyl isomerase,
cyclophilin A (Cyp A) that is incorporated into the virion by
binding to CA and its presence enhances viral infectivity
[80,81]. Actin, and various actin-binding proteins have also
been detected [82]. The fate of these proteins after entering
the cells is mostly unknown, but many were already
fragmented in the virions, and the involvement of the PR at
least in degrading elongation factor-1 alpha (EF1α) was
demonstrated [82]. During in vitro incubation of EIAV cores
in the presence of a chelating agent (EDTA) the NC was
further processed in situ into smaller fragments by the
incorporated PR [70,83]. To extend our initial studies of
EIAV NC cleavage, we used short synthetic peptides derived
from the highly homologous HIV-1 and HIV-2 NC as
substrates for PR. Based on these studies we predicted that
the cleavage site located in the proximal (N-terminal zinc
finger) of HIV-1 NC should be between Phe and Asn (see
Fig. 2B), shifted one residue downstream compared to the
EIAV cleavage site [84]. This was later confirmed using NC
protein as a substrate [85]. Among the other proteins of the
core, the HIV-1 RT, RNase H and Nef have been
demonstrated to be substrates of the HIV-1 PR [71,86,87].
Furthermore, the PR itself undergoes self-degradation [88],
while Vpr, a nucleocytoplasmic shuttling protein of the PIC
[89], remains intact in the capsid [71]. We have observed
that the capsid protein is also a substrate of the PR in an
acidic environment, while the integrase, an essencial
component of the PIC is resistant towards proteolysis [32].
New cleavage sites in CA were identified (Fig. 2B), and one
of them showed a special pH sensitivity, as explained in
comparison with the Gag-Pro-Pol cleavage sites (see above).
The protease-mediated cleavage of viral and cellular proteins
of the core may be important for the formation of proper
preintegration complex.
4.1.3. Effect of HIV Protease Inhibitors in the Early Phase
of Viral Replication Cycle
Several research groups, including ours, found an
inhibitory effect of HIV-1 protease inhibitors on the early
phase of viral replication [90-94], while others did not
observe such an effect [95-97]. Action of the PR inhibitors in
the early-phase may be one factor contributing to the
enhanced in vivo antiviral potency of these drugs (for
example [98]). The different trafficking route of HIV-1
depending on the presence of receptor and virus isolate [68]
may be responsible at least in part for the conflicting results
obtained with PR inhibitors to study the role of PR in the
early phase of the viral life-cycle. Further studies are
required to clarify the role of the virally coded PR in the
early phase of viral replication. Furthermore, PR inhibitors
targeting the early-phase effect on the endocytotic route may
need somewhat different characteristics from the currently
used inhibitors. Those PR inhibitors which can efficiently
interfere with both the early and late phases of the viral life-
cycle would have a potentially great advantage: besides
protecting cells from infection, they can also influence the
chronically infected (integrated viral genome-containing)
cells, while the RT and IN inhibitors do not affect
chronically infected cells. However, at the present stage, we
cannot exclude the possibility that the PR-mediated
proteolysis of viral proteins in the early phase, if it occurs in
vivo, is not favoring the infection; rather it is a “suicidal”
effect.
4.2. Involvement of the Proteasome System in the
Cellular Defense Against HIV-1 Infection
Proteasomes are large multidomain complexes, which are
involved in the ATP-dependent degradation of most of the
intracellular proteins [99]. It has been shown that
proteasomal inhibitors, acting in the early phase of the life-
cycle, increased the efficiency of HIV infection, presumably
by preventing the degradation of the proteins of infectious
virions [100]. The substrates for proteasomal degradation
appear to be complexes containing largely completed reverse
transcription products, or fully formed preintegration
complexes [101].
ACKNOWLEDGEMENTS
We wish to thank Dr. Péter Bagossi and Gábor Zahuczky
for preparing figures and for valuable discussions. We are
grateful to Maritta P. Grau for editorial assistance. Our
research described in this review was sponsored in part by
the Hungarian Science and Research Fund (OTKA T 30092
and T43482).
ABBREVIATIONS
AIDS = Acquired immunodeficiency syndrome
CA = Capsid protein
Cyp A = Cyclophilin A
EF1α= Elongation factor-1 alpha
EIAV = Equine infectious anemia virus
HAART = Highy active antiretroviral therapy
HIV = Human immunodeficiency virus
IN = Integrase
MA = Matrix protein
MuLV = Murine leukemia virus
1812 Current Pharmaceutical Design, 2003, Vol. 9, No. 22 Tözsér and Oroszlan
NC = Nucleocapsid protein
PACE4 = Paired amino acids-cleaving enzyme 4
PC = Prohormone convertase
PIC = Preintegration complex
PR = Viral protease
RNase H = Ribonuclease H
RT = Reverse transcriptase
SIV = Simian immunodeficiency virus
SU = Surface protein
TM = Transmembrane protein
REFERCENCES
References 102-104 are related articles recently published in
Current Pharmaceutical Design.
[1] Temesgen Z. Current status of antiretroviral therapies.
Expert Opin Pharmacother 2001; 2: 1239-46.
[2] Telenty A, Paolo Rizzardi G. Limits to potent antiretroviral
therapy. Rev Med Virol 2000; 10: 385-93.
[3] Carr A, Cooper DA. Adverse effects of antiretroviral
therapy. Lancet 2000; 356: 1423-30.
[4] Tözsér J. HIV inhibitors: problems and reality. Ann N Y
Acad Sci 2001; 946: 145-59.
[5] Moyle G, Carr A. HIV-associated lipodystrophy, metabolic
complications, and antiretroviral toxicities. HIV Clin Trials
2002; 3: 89-98.
[6] Parker SD, Hunter E. Activation of the Mason-Pfizer
monkey virus protease within immature capsids in vitro.
Proc Natl Acad Sci USA 2001; 98: 14631-6.
[7] Davis DA, Brown CA, Newcomb FM, Boja ES, Fales HM,
Kaufman J, et al. Reversible oxidative modification as a
mechanism for regulating retroviral protease dimerization
and activation. J Virol 2003; 77: 3319-25.
[8] Kubota S, Oroszlan S. The role of redox signaling in the
processing of Gag polyprotein in immature retrovirus. In:
Pasquier C. Ed, XI Biennial Meeting of the Society for Free
Radical Research International (Paris, France, July 16-20,
2002). Monduzzi Editore Italy 2003; 193-7.
[9] Oroszlan S, Luftig RB. Retroviral proteinases. Curr Topics
Microbiol Immunol 1990; 157: 153-85.
[10] Wlodawer A. Rational approach to AIDS drug design
through structural biology. Annu Rev Med 2002; 53: 595-
614.
[11] Vondrasek J, W lodawer A. HIVdb: a database of the
structures of human immunodeficiency virus protease.
Proteins 2002; 49: 429-31.
[12] Szeltner Z, Polgár L. Rate-determining steps in HIV-1
protease catalysis. The hydrolysis of the most specific
substrate. J Biol Chem 1996; 271: 32180-4.
[13] Tomasselli AG, Heinrikson RL. Specificity of retroviral
proteinases: an analysis of viral and nonviral protein
substrates. Methods Enzymol 1994; 241: 279-301.
[ 14] Dunn BM, Gustchina A, Wlodawer A, Kay J. Subsite
preferences of retroviral proteinases. Methods Enzymol
1994; 241: 254-78.
[15] Tözsér J. Specificity of retroviral proteinases based on
substrates containing tyrosine and proline at the site of
cleavage. Pathol Oncol Res 1997; 3: 141-6.
[16] Louis JM, Weber IT, Tözsér J, Clore GM, Gronenborn AM.
HIV-1 protease: maturation, enzyme specificity and drug
resistance. Adv Pharmacol 2000; 49: 111-46.
[17] Pettit SC, Simsic J, Loeb DD, Everitt L, Huchinson CA 3rd,
Swanstrom R. Analysis of retroviral protease cleavage sites
reveals two types of cleavage sites and the structural
requirements of the P1 amino acid. J Biol Chem 1991; 266:
14539-47.
[18] Griffiths JT, Phylip LH, Konvalinka J, Strop P, Gustchina
A, Wlodawer A, et al. Different requirements for productive
interaction between the active site of HIV-1 proteinase and
substrates containing -hydrophobic*hydrophobic- or -
aromatic*Pro- cleavage sites. Biochemistry 1992; 31: 5193-
200.
[19] Tözsér J, Weber IT, Gustchina A, Bláha I, Copeland TD,
Louis JM, et al. Kinetic and modeling studies of S3-S3'
subsites of HIV proteinases. Biochemistry 1992; 31: 4794-
800.
[20] Schechter I, Berger A. On the size of the active site in
proteases. I. papain. Biochem Biophys Res Commun 1967;
27: 157-162.
[21] Oroszlan S, Henderson LE, Stephenson JR, Copeland TD,
Long CW, Ihle, JN et al. Amino- and carboxyl-terminal
amino acid sequences of proteins coded by gag gene of
murine leukemia virus. Proc Natl Acad Sci USA 1978; 75:
1404-8.
[22] Copeland TD, Wondrak EM, Tözsér J, Roberts MM,
Oroszlan S. Substitution of proline with pipecolic acid at
the scissile bond converts a peptide substrate of HIV
proteinase into a selective inhibitor. Biochem Biophys Res
Commun 1990; 169: 310-4.
[23] MacArthur WM, Thornton JM. Influence of proline
residues on protein conformation. J Mol Biol 1990; 218:
397-412.
[24] Vance JE, LeBlanc DA, Wingfield P, London RE.
Conformational selectivity of HIV-1 protease cleavage of
X-Pro peptide bonds and its implications. J Biol Chem
1997; 72: 15603-6.
[25] McCornack MA, Kakalis LT, Castera C, Handschumacher
RE, Armitage IM. HIV protease substrate conformation:
modulation by cyclophilin A. FEBS Lett 1997; 414, 84-8.
[26] Tözsér J, Bagossi P, Weber IT, Louis JM, Copeland TD,
Oroszlan S. Studies on the symmetry and sequence context
dependence of the HIV-1 proteinase specificity. J Biol
Chem 1997; 272: 16807-14.
[27] Korber B, Kuiken C, Foley B, Hahn B, McCuthan F,
Mellors J. et al. Eds, Human Retroviruses and AIDS. Los
Alamos, NM, USA, 1998.
[28] Pettit SC, Moody MD, Wehbie RS, Kaplan AH, Nantermet
PV, Klein, CA, et al. The p2 domain of human
immunodeficiency virus type 1 Gag regulates sequential
proteolytic processing and is required to produce fully
infectious virions. J Virol 1994; 68: 8017-27.
[29] Tong L, Pav S, Pargellis C, Dô F, Lamarre D, Anderson
PC. Crystal structure of human immunodeficiency virus
(HIV) type 2 protease in complex with a reduced amide
inhibitor and comparison with HIV-1 protease structures.
Proc Natl Acad Sci USA 1993; 90: 8387-91.
[30] Weber IT, Wu J, Adomat J, Harrison RW, Kimmel AR,
Wondrak EM et al. Crystallographic analysis of human
immunodeficiency virus 1 protease with an analog of the
conserved CA-p2 substrate. Interactions with frequently
occurring glutamic acid residue at P2' position of substrates.
Eur J Biochem 1997; 249: 523-30.
Proteolytic Events of HIV-1 Replication Current Pharmaceutical Design, 2003, Vol. 9, No. 22 1813
[31] Boross P, Bagossi P, Copeland TD, Oroszlan S, Louis JM,
Tözsér J. Effect of substrate residues on the P2’ preference
of retroviral proteinases. Eur J Biochem 1999; 264: 921-9.
[32] Tözsér, J., Shulenin, S., Kádas, J., Boross, P., Copeland,
T.D., Nair, M.G. et al. Human immunodeficiency virus
type-1 capsid protein is a substrate of retroviral proteinase
while integrase is resistant toward proteolysis. Virology
2003; in press.
[33] Sham HL, Kempf DJ, Molla A, Marsh KC, Kumar GN,
Chen CM, et al. ABT-378, a highly potent inhibitor of the
human immunodeficiency virus protease. Antimicrob
Agents Chemother 1998; 42: 3218-24.
[34] Doyon L, Croteau G, Thibeault D, Poulin F, Pilote L,
Lamarre D. Second locus involved in human
immunodeficiency virus type 1 resistance to protease
inhibitors. J Virol 1996; 70: 3763-9.
[35] Croteau G, Doyon L, Thibeault D, McKercher G, Pilote L,
Lamarre D. Impaired fitness of human immunodeficiency
virus type 1 variants with high-level resistance to protease
inhibitors. J Virol 1997; 71: 1089-96.
[36] Zhang YM, Imamichi H, Imamichi T, Lane HC, Falloon J,
Vasudevachari MB, et al. Drug resistance during indinavir
therapy is caused by mutations in the protease gene and in
its Gag substrate cleavage sites. J Virol 1997; 71: 6662-
6670.
[37] Côté HC, Brumme ZL, Harrigan PR. Human immunodefi-
ciency virus type 1 protease cleavage site mutations
associated with protease inhibitor cross-resistance selected
by indinavir, ritonavir, and/or saquinavir. J Virol 2001; 75:
589-94.
[38] Robinson LH, Myers RE, Snowden BW, Tisdale M, Blair
ED. HIV type 1 protease cleavage site mutations and viral
fitness: implications for drug susceptibility phenotyping
assays. AIDS Res Hum Retroviruses 2000; 16: 1149-56.
[39] Tözsér J, Bláha I, Copeland TD, Wondrak EM, Oroszlan S.
Comparison of the HIV-1 and HIV-2 proteinases using
oligopeptide substrates representing cleavage sites in Gag
and Gag-Pol polyproteins. FEBS Lett 1991; 281, 77-80.
[ 40] W ondr a k EM, L ouis JM, de Roc quigny H, Che rma nn JC,
Roque s BP. The gag precursor contains a specific HIV-1
protease cleavage site between the NC (P7) and P1 proteins.
FEBS Lett. 1993; 333: 21-24.
[41] Fehér A, Weber IT, Bagossi P, Boross P, Mahalingam B,
Louis JM, et al. Effect of sequence polymorphism and drug
resistance on two HIV-1 Gag processing sites. Eur J
Biochem 2002; 269: 4114-20.
[42] Pettit SC, Sheng N, Tritch R, Erickson-Viitanen S,
Swanstrom R. The regulation of sequential processing of
HIV-1 Gag by the viral protease. Adv Exp Med Biol 1998;
436: 15-25.
[43] Doms RW, Moore JP. HIV-1 membrane fusion: targets for
opportunity. J Cell Biol 2000; 151: 9-13.
[44] Clapham PR, McKnight A. Cell surface receptors, virus
entry and tropism of primate lentiviruses. J Gen Virol 2002;
83: 1809-29.
[45] Moulard M, Decroly E. Maturation of HIV envelope
glycoprotein precursors by cellular endoproteases. Biochem
Biophys Acta 2000; 1469: 121-32.
[46] McCune JM, Rabin LB, Feinberg MB, Leiberman M,
Mosek JC, Reyes GR, et al. Endoproteolytic cleavage of
gp160 is required for the activation of human
immunodeficiency virus. Cell 1989; 53: 55-67.
[47] Kido H, Kamoshita K, Fukutomi A, Katunuma N.
Processing protease for gp160 human immunodeficiency
virus type 1 envelope glycoprotein precursor in human T4+
lymphocytes. Purification and characterization. J Biol Chem
1993; 268: 13406-13.
[48] Bendjennat M, Bahbouhi B, Bahraoui E. Purification and
characterization of a Ca2+-independent endoprotease
activity from peripheral blood lymphocytes: involvement in
HIV-1 gp160 maturation. Biochemistry 2001; 40: 4800-10.
[49] Hallenberger S, Bosch V, Angliker H, Shaw E, Klenk HD,
Garten W. Inhibition of furin-mediated cleavage activation
of HIV-1 glycoprotein gp160. Nature 1992; 360: 358-61.
[50] Anderson ED, Thomas L, Hayflick JS, Thomas G.
Inhibition of HIV-1 gp160-dependent membrane fusion by
a furin directed alpha 1-antitrypsin variant. J Biol Chem
1993; 268: 24887-91.
[51] Clements GJ, Price-Jones MJ, Stephens PE, Sutton C,
Schulz TF, Clapham PR, et al. The V3 loops of the HIV-1
and HIV-2 surface glycoproteins contain proteolytic
cleavage sites: a possible function in viral fusion? AIDS
Res Hum Retroviruses 1991; 7: 3-16.
[52] Niwa Y, Yano M, Futaki S, Okumura Y, Kido H. T-cell
membrane-associated serine protease, tryptase TL2, binds
human immunodeficiency virus type 1 gp120 and cleaves
the third-variable-domain loop of gp120. Neutralizing
antibodies of human immunodeficiency virus type 1 inhibit
cleavage of gp120. Eur J Biochem 1996; 237: 64-70.
[53] Handley MA, Steigbigel RT, Morrison SA. A role for
urokinase-type plasminogen activator in human
immunodeficiency virus type 1 infection of macrophages. J
Virol 1996; 70: 4451-6.
[54] Henderson LE, Copeland TD, Sowder RC, Schultz AM,
Oroszlan S. Analysis of proteins and peptides purified from
sucrose gradient banded HTLV-III. In: Bolognesi D Ed,
Human Retroviruses, Cancer, and AIDS: Approaches to
prevention and therapy. Alan R Liss Inc New York 1988;
135-47.
[55] Ott DE, Core n L V, Cope land TD, K ane BP, Johnson DG ,
Sowder RC 2nd, et a l. Ubiquitin is covalently attached to the
p6Gag proteins of human immunodeficiency virus type 1
and simian immunodeficiency virus and to the p12Gag
protein of Moloney murine leukemia virus. J Virol 1998; 72:
2962-8.
[56] Ott DE, Coren LV, Sowder RC 2nd, Adams J, Nagashima
K, Schubert U. Equine infectious anemia virus and the
ubiquitin-proteasome system. J Virol 2002; 76, 3038-44.
[57] Putterman D, Pepinsky RB, Vogt VM. Ubiquitin in avian
leukosis virus particles. Virology 1990; 176: 633-7.
[58] Schubert U, Ott DE, Chertova EN, Welker R, Tessmer U,
Princiotta M, et al. Proteasome inhibition interferes with
gag polyprotein processing, release, and maturation of HIV-
1 and HIV-2. Proc Natl Acad Sci USA 2000; 97: 13057-62.
[59] Shubert U, Anton LC, Gibbs J, Norbury C, Yewdell JW,
Bennink JR. Rapid degradation of a large fraction of newly
synthesized proteins by proteasomes. Nature 2000; 404:
770-4.
[60] Kisselev AF, Goldberg AL. Proteasome inhibitors: from
research tools to drug candidates. Chem Biol 2001; 8: 739-
58.
[61] Schmidtke G, Holzhutter HG, Bogyo M, Kairies N, Groll
M, de Giuli R, et al. How an inhibitor of HIV-1 protease
modulates proteasome activity. J Biol Chem 1999; 274:
35734-40.
[62] Pajonk F, Himmelsbach J, Riess K, Sommer A, McBride
WH. The human immunodeficiency virus (HIV)-1 protease
1814 Current Pharmaceutical Design, 2003, Vol. 9, No. 22 Tözsér and Oroszlan
inhibitor saquinavir inhibits proteasome function and causes
apoptosis and radiosensitization in non-HIV-associated
human cancer cells. Cancer Res 2002; 62: 5230-5.
[63] Marsh M, Pelchen-Matthews A. Endocytosis in viral
replication. Traffic 2000; 1: 525-32.
[64] Katen LJ, Januszeski MM, Anderson WF, Hasenkrug KJ,
Evans LH. Infectious entry by amphotropic as well as
ecotropic murine leukemia viruses occurs through an
endocytic pathway. J Virol 2001; 75: 5018-26.
[65] Grewe C, Beck A, Gelderblom HR. HIV: early virus-cell
interactions. J Acquir Immune Defic Syndr 1990; 3: 965-
74.
[66] Maréchal V, Clavel F, Heard JM, Schwartz O. Cytosolic
Gag p24 as an index of productive entry of human
immunodeficiency virus type 1. J Virol 1998; 72: 2208-12.
[67] Schaeffer E, Geleziunas R, Green WC. Human
immunodeficiency virus type 1 Nef functions at the level of
virus entry by enhancing cytoplasmic delivery of virions. J
Virol 2001; 75: 2993-3000.
[68] Fackler OT, Peterlin BM. Endocytic entry of HIV-1. Curr
Biol 2000; 10: 1005-8.
[69] Maréchal V, Prevost MC, Petit C, Perret E, Heard JM,
Schwartz O. Human immunodeficiency virus type 1 entry
into macrophages mediated by macropinocytosis. J Virol
2001; 75: 11166-77.
[70] Roberts MM, Oroszlan S. The preparation and biochemical
characterization of intact capsids of equine infectious
anemia virus. Biochem Biophys Res Commun 1989; 160:
486-94.
[71] Welker R, Hohenberg H, Tessmer U, Huckhagel C,
Krausslich HG (2000) Biochemical and structural analysis
of isolated mature cores of human immunodeficiency virus
type 1. J Virol 2000; 74: 1168-77.
[72] Shoeman RL, Höner B, Stoller TJ, Kesselmeier C, Miedel
MC, Traub P, et al. Human immunodeficiency virus type 1
protease cleaves the intermediate filament proteins
vimentin, desmin, and glial fibrillary acidic protein. Proc
Natl Acad Sci USA 1990; 87: 6336-40.
[73] Shoeman RL, Sachse C, Höner B, Mothes E, Kaurmann M,
Traub P. Cleavage of human and mouse cytoskeletal and
sarcomeric proteins by human immunodeficiency virus type
1 protease. Actin, desmin, myosin and tropomyosin. Am J
Pathol 1993; 142: 221-30.
[74] Thomas EK, Connelly RJ, Pennathur S, Dubrovsky L,
Haffar OK, Bukrinsky MI. Anti-idiotypic antibody to the
V3 domain of gp120 binds to vimentin: a possible role of
intermediate filaments in the early steps of HIV-1 infection
cycle. Viral Immunol 1996; 9: 73-87.
[75] Tözsér J, Bagossi P, Boross P, Louis JM, Majerova E,
Oroszlan S, et al. Effect of serine and tyrosine
phosphorylation on retroviral proteinase substrates. Eur J
Biochem 1999; 265: 423-9.
[76] Bukrinskaya A, Brichacek B, Mann A, Stevenson M.
Establishment of a functional human immunodeficiency
virus type 1 (HIV-1) reverse transcription complex involves
the cytoskeleton. J Exp Med 1998; 188: 2113-25.
[77] Riviére Y, Blank V, Kourilsky P, Israël A. Processing of
the precursor of NF-κB by the HIV-1 protease during acute
infection. Nature 1991; 350, 625-6.
[78] Kaplan AH, Swanstrom R. Human immunodeficiency virus
type 1 Gag proteins are processed in two cellular
compartments. Proc Natl Acad Sci USA 1991; 88, 4528-32.
[79] Ohlmann T, Prévôt D, Décimo D, Roux F, Garin J, Morley
SJ, et al. In vitro cleavage of eIF4GI but not eIF4GII by
HIV-1 protease and its effects on translation in the rabbit
retikulocyte lysate system. J Mol Biol 2002; 318: 9-20.
[80] Franke EK, Yuan HE, Luban J. Specific incorporation of
cyclophylin A into HIV-1 virions. Nature 1994; 372: 359-
62.
[81] Thali M, Bukovsky A, Kondo E, Rosenwirth B, Walsh CT,
Sodorski J, et al. Functional association of cyclophilin A
with HIV-1 virions. Nature 1994; 372: 363-5.
[82] Ott DE, Coren LV, Johnson DG, Kane BP, Sowder RC 2nd,
Kim YD, et al. Actin-binding cellular proteins inside human
immunodeficiency virus 1. Virology 2000; 266: 42-51.
[83] Roberts MM, Copeland TD, Oroszlan S. In situ processing
of a retroviral nucleocapsid protein by the viral proteinase.
Protein Eng 1991; 4: 695-700.
[84] Tözsér J, Friedman D, Weber IT, Bláha I, Oroszlan S.
Studies on the substrate specificity of the proteinase of
equine infectious anemia virus using oligopeptide
substrates. Biochemistry 1993; 32: 3347-53.
[85] Wondrak EM, Sakaguchi K, Rice WG, Kun E, Kimmel AR,
Louis JM. Removal of zinc is required for processing of the
mature nucleocapsid protein of human immunodeficiency
virus, type 1, by the viral protease. J Biol Chem 1994; 269:
21948-50.
[86] Tomasselli AG, Sarcich JL, Barrett LJ, Reardon IM, Howe
WJ, Evans DB, et al. Human immunodeficiency virus type-
1 reverse transcriptase and ribonuclease H as substrates of
the viral protease. Protein Sci 1993; 2: 2167-76.
[87] Welker R, Kottler H, Kalbitzer HR, Krausslich HG. Human
immunodeficiency virus type 1 Nef protein is incorporated
into virus particles and specifically cleaved by the viral
proteinase. Virology 1996; 219: 228-36.
[88] Mildner AM, Rothrock DJ, Leone JW, Bannow CA, Lull
JM, Reardon IM, et al. The HIV-1 protease as enzyme and
substrate: mutagenesis of autolysis sites and generation of a
stable mutant with retained kinetic properties. Biochemistry
1994; 33: 9405-13.
[89] Sherman MP, de Noronha CM, Heusch MI, Greene S.,
Greene WC. Nucleocytoplasmic shuttling by human
immunodeficiency virus type 1 Vpr. J Virol 2001; 75:
1522-32.
[90] Baboonian C, Dalgleish A, Bountiff L, Gross J, Oroszlan S,
Rickett G, et al. HIV-1 proteinase is required for synthesis
of pro-viral DNA. Biochem Biophys Res Commun 1991;
179: 17-24.
[91] Venaud S, Yahi N, Fehrentz JL, Guettari N, Nisato D,
Hirsch I, et al. Inhibition of HIV by an anti-HIV protease
synthetic peptide blocks an early step of viral replication.
Res Virol 1992; 143, 311-9.
[92] Nagy K, Young M, Baboonian C, Merson J, Whittle P,
Oroszlan S. Antiviral activity of human immunodeficiency
virus type 1 protease inhibitors in a single cycle of
infection: evidence for a role of protease in the early phase.
J Virol 1994; 68: 757-65.
[93] Panther LA, Coombs RW, Aung SA, dela Rosa C, Gretch
D, Corey L. Unintegrated HIV-1 circular 2-LTR proviral
DNA as a marker of recently infected cells: Relative effect
of recombinant CD4, zidovudine and saquinavir in vitro. J
Med Virol 1999; 58: 165-73.
[94] Goto T, Nakano T, Kohno T, Morimatsu S, Morita C, Hong
W, et al. Targets of a protease inhibitor, KNI-272, in HIV-
1-infected cells. J Med Virol 2001; 63: 203-9.
[95] Jacobsen H, Ahlborn-Laake J, Gugel R, Mous J.
Progression of early steps of human immunodeficiency
Proteolytic Events of HIV-1 Replication Current Pharmaceutical Design, 2003, Vol. 9, No. 22 1815
virus type 1 replication in the presence of an inhibitor of
viral protease. J Virol 1992; 66: 5087-92.
[96] Kaplan AH, Manchester M, Smith T, Yang YL, Swanstrom
R. Conditional human immunodeficiency virus type 1
protease mutants show no role for the viral protease early in
virus replication. J Virol 1996; 70, 5840-4.
[97] Uchida H, Maeda Y, Mitsuya H. HIV-1 protease does not
play a critical role in the early phase of HIV-1 infection.
Antiviral Res 1997; 36, 107-13.
[98] Collier AC, Coombs RW, Schoenfeld DA, Bassett RL,
Timpone J, Baruch A. et al. Treatment of human
immunodeficiency virus infection with saquinavir,
zidovudine, and zalcitabine. AIDS Clinical Trials Group. N
Engl J Med 1996; 334: 1011-7.
[99] Voges D, Zwickl P, Baumaister W. The 26S proteasome: a
molecular machine designed for controlled proteolysis.
Annu Rev Biochem 1999; 68: 1015-68.
[100] Schwartz O, Maréchal V, Friguet B, Arenzana-Seisdedos F,
Heard JM. Antiviral activity of the proteasome on incoming
human immunodeficiency virus type 1. J Virol 1998; 72:
3845-50.
[101] Butler SL, Johnson EP, Bushman FD. Human
immunodeficiency virus cDNA metabolism: notable
stability of two-long terminal repeat circles. J Virol 2002;
76: 3739-47.
[102] Campiani G, Ramunno A, Maga G, Nacci V, Fattorusso C,
Catalanotti B, et al. Non-nucleoside HIV-1 reverse
transcriptase (RT) inhibitors: past, present, and future
perspectives. Curr Pharm Des 2002; 8(8): 615-57.
[103] Jiang S, Zhao Q, Debnath AK. Peptide and non-peptide
HIV fusion inhibitors. Curr Pharm Des 2002; 8(8): 563-80.
[104] Tarrago-Litvak L, Andreola ML, Fournier M, Nevinsky
GA, Parissi V, de Soultrait VR, et al. Inhibitors of HIV-1
reverse transcriptase and integrase: classical and emerging
therapeutical approaches. Curr Pharm Des 2002; 8(8): 595-
614.
... The role of the protease (PR) of human retroviruses in the late phase of virus replication by processing the Gag and Gag-Pol polyproteins has been well established (for a review, see [1]). The viral polyprotein is translated from different open reading frames (ORFs), and the Gag polyprotein consists of the major structural proteins: the matrix (MA), the capsid (CA), the nucleocapsid (NC), and the p6 at the C terminus. ...
... Based on the above-mentioned findings, the potential role of the PR in the early phase of the retroviral life-cycle was suggested, either by performing post-maturation cleavages of NC and CA or by cleaving protein substrates [1]. As compared to the function in the late phase events, the role of the PR in the early phase is less well established and is still Peptides representing the predicted cleavage sites in the second zinc-fingers were not substrates of the HIV-1 PR [8]; however, in vitro studies indicated another site of cleavage in the second zinc-finger [9]. ...
... Based on the above-mentioned findings, the potential role of the PR in the early phase of the retroviral life-cycle was suggested, either by performing post-maturation cleavages of NC and CA or by cleaving protein substrates [1]. As compared to the function in the late phase events, the role of the PR in the early phase is less well established and is still controversial. ...
Article
Full-text available
To explore the sequence context-dependent nature of the human immunodeficiency virus type 1 (HIV-1) protease’s specificity and to provide a rationale for viral mutagenesis to study the potential role of the nucleocapsid (NC) processing in HIV-1 replication, synthetic oligopeptide substrates representing the wild-type and modified versions of the proximal cleavage site of HIV-1 NC were assayed as substrates of the HIV-1 protease (PR). The S1′ substrate binding site of HIV-1 PR was studied by an in vitro assay using KIVKCF↓NCGK decapeptides having amino acid substitutions of N17 residue of the cleavage site of the first zinc-finger domain, and in silico calculations were also performed to investigate amino acid preferences of S1′ site. Second site substitutions have also been designed to produce “revertant” substrates and convert a non-hydrolysable sequence (having glycine in place of N17) to a substrate. The specificity constants obtained for peptides containing non-charged P1′ substitutions correlated well with the residue volume, while the correlation with the calculated interaction energies showed the importance of hydrophobicity: interaction energies with polar residues were related to substantially lower specificity constants. Cleavable “revertants” showed one residue shift of cleavage position due to an alternative productive binding mode, and surprisingly, a double cleavage of a substrate was also observed. The results revealed the importance of alternative binding possibilities of substrates into the HIV-1 PR. The introduction of the “revertant” mutations into infectious virus clones may provide further insights into the potential role of NC processing in the early phase of the viral life-cycle.
... The role of the mature homodimeric PR is the limited proteolysis of the polyproteins into functional subunits, and the intradomain bonds are cleaved at specific sites with different cleavage rates. The function of HIV-1 PR is essential for the viral infectivity; therefore, the treatment of acquired immunodeficiency syndrome (AIDS) includes the use of inhibitors that block its activity [25,26]. ...
... Our research group, the Laboratory of Retroviral Biochemistry was established at the University of Debrecen in 1992 by József Tőzsér, after working together with Dr. Oroszlan in the Molecular Virology and Carcinogenesis Laboratory at NCI-Frederick Center for Cancer Research. Most studies of our research group followed his ideas, including the investigation of HIV-1 and other retroviral proteases, antiviral inhibitors and enzyme specificity, works which have been covered by reviews [25,27,30]. Several studies of our research group-including the present work-were inspired by the specificity studies of our laboratory performed previously together with Dr. Oroszlan and his colleagues. ...
Article
Full-text available
Proteolytic enzymes have great significance in medicine and the pharmaceutical industry and are applied in multiple fields of life sciences. Therefore, cost-efficient, reliable and sensitive real-time monitoring methods are highly desirable to measure protease activity. In this paper, we describe the development of a new experimental approach for investigation of proteolytic enzymes. The method was designed by the combination of recombinant fusion protein substrates and bio-layer interferometry (BLI). The protease (PR) of human immunodeficiency virus type 1 (HIV-1) was applied as model enzyme to set up and test the method. The principle of the assay is that the recombinant protein substrates immobilized to the surface of biosensor are specifically cleaved by the PR, and the substrate processing can be followed by measuring change in the layer thickness by optical measurement. We successfully used this method to detect the HIV-1 PR activity in real time, and the initial rate of the signal decrease was found to be proportional to the enzyme activity. Substrates representing wild-type and modified cleavage sites were designed to study HIV-1 PR’s specificity, and the BLI-based measurements showed differential cleavage efficiency of the substrates, which was proven by enzyme kinetic measurements. We applied this BLI-based assay to experimentally confirm the existence of extended binding sites at the surface of HIV-1 PR. We found the measurements may be performed using lysates of cells expressing the fusion protein, without primary purification of the substrate. The designed BLI-based protease assay is high-throughput-compatible and enables real-time and small-volume measurements, thus providing a new and versatile approach to study proteolytic enzymes.
... A pillar among N-heterocycles is β-lactams [8,9], also referred as 2azetidinones, which have seen unparalleled success rate as antibacterial agents such as penicillin, cephalosporins, carbapenems, nocardicins, monobactams, clavulanic acid, and others [10][11][12]. Although initially the majority of the βlactams were developed as antibiotics, later they were extended to other biological applications such as inhibiting human tryptase & cholesterol absorption, as antimicrobials, anticancer agents, antidiabetics, as anti-inflammatory agents, antiparkinsonian and anti-HIV agents [13][14][15][16][17][18][19][20]. ...
Article
Full-text available
An efficient synthetic strategy was developed for the synthesis of hybrid pharmacophores; encompass the merging of β-lactams, 1,8-naphthyridine and secondary amines/pyridines. With a simple unified synthetic protocol, we prepared a series of substituted 3-chloro-1-((3-(2-(trifluoromethyl)phenyl)-1,8-naphthyridin-2-yl)amino)azetidin-2-one compounds (8a–8j) and evaluated for their antimicrobial, and anticancer activities (against MDA-MB-231 cell line). Notably, compounds 8d, 8g, and 8j were identified as potent anticancer molecules and comparable to Cisplatin. Compounds 8d and 8g were found to have promising antitubercular efficacy even greater than the standards Streptomycin and Ciprofloxacin. Compounds 8a, 8d, 8g, 8i, and 8j also displayed potency against both Gram-positive and Gram-negative bacteria and comparable to Ampicillin & Ciprofloxacin. Further, the biological activities were supported and studied by DFT studies and ADMET predictions. Graphical Abstract
... Earlier results strongly suggested that appropriate stability of the core also had a main role in HIV-1 infection [2-7] influencing the decapsidation process in the early phase where the exact identity of the factors involved is still not fully resolved. Besides the presence of various viral proteins, the protease (PR) is also part of the core [8] and, therefore, it might contribute to the viral infectivity by further processing viral proteins or by cleaving cellular ones [9]. At the late phase of the viral life cycle, proteolytic cleavage of Gag and Gag-Pol polyproteins is essential for the production of the mature, infectious virus [10]. ...
Article
Full-text available
The capsid protein of the human immunodeficiency virus type 1 has been found to be a substrate of the retroviral protease in vitro, and its processing was predicted to be strongly dependent on a pH-induced conformational change. Several protease cleavage sites have been identified within the capsid protein, but the importance of its cleavage by the viral protease at the early phase of infection is controversial. To confirm the relevance of this process, we aimed to design, produce, and characterize mutant capsid proteins, in which the protein susceptibility toward HIV-1 protease is altered without affecting other steps of the viral life cycle. Our results indicate that while the introduced mutations changed the cleavage rate at the mutated sites of the capsid protein by HIV-1 protease, some of them caused only negligible or moderate structural changes (A78V, L189F, and L189I). However, the effects of other mutations (W23A, A77P, and L189P) were dramatic, as assessed by secondary structure determination or cyclophilin A-binding assay. Based on our observations, the L189F mutant capsid remains structurally and functionally unchanged and may therefore be the best candidate for use in studies aimed at better understanding the role of the protease in the early postentry events of viral infection or retrovirus-mediated gene transduction.
... The endopeptidase, leupeptin, differs from most other natural protease inhibitors because it contains an acetyl group and possesses an arginal aldehyde in place of the more common terminal carbonyl. Unlike most of its synthetic protease counterparts, leupeptin is believed to be much less toxic to humans and has generated considerable interest as a possible therapeutic intervention for purposes as diverse as the treatment of hearing loss (10 μM -100 μM) [21,22], treatment of malaria [23], HIV [24,25],, or anywhere protease inhibition may be otherwise indicated [26]. Direct exposure of organogenesis-stage rat and mouse conceptuses grown in whole embryo culture (rWEC) to leupeptin (10 μM -100 μM) resulted in a significant accumulation of undigested protein in the lysosomes (~80% increase) and the significant reduction of growth and widespread dysmorphogenesis [17]. ...
Article
The process of embryonic nutrition in rodent conceptuses during organogenesis has been shown to involve a dominant histiotrophic mechanism where essential developmental substrates and micronutrients are supplied as whole maternal proteins or cargoes associated with proteins. The histiotrophic nutrition pathways (HNP) responsible for uptake and initial processing of proteins across maternal-conceptal interfaces involve uptake via receptor mediated endocytosis and protein degradation via lysosomal proteolysis. Chemical inhibition of either process can lead to growth deficits and malformation in the embryo (EMB), but selective inhibition of either HNP component will elicit a different subset of developmental perturbations. In vitro, whole embryo culture exposure of GD10 or GD11 rat conceptuses to the natural protease inhibitor, leupeptin, leads to significant reductions in all measured embryonic growth parameters as well as a myriad of other effects. Leupeptin doses of 10 μM or 20 μM over a 26-h period (GD10-GD11) and 50 μM over a 3 h pulse period produced significant decreases in the clearance of FITC-albumin from culture media. The near complete loss of acid soluble fluorescence and increased total visceral yolk sac (VYS) protein content confirmed the selective inhibition of proteolysis. Inhibition of lysosomal proteolysis thus deprives the developing EMB of essential nutrient amino acids producing conditions akin to amino acid starvation, but may also cause direct effects on pathways critical for normal growth and differentiation. Following leupeptin exposure for 26 or 6 h, total glutathione (GSH) concentrations dropped significantly in the VYS, but only slightly in yolk sac (YSF) and amniotic (AF) fluids. Cys concentrations increased in VYS and EMB, but dropped in YSF and AF fluids. Redox potentials (Eh) for the glutathione disulfide (GSSG)/glutathione (GSH) redox couple trended significantly toward the positive, confirming the net oxidation of conceptual tissues following leupeptin treatment. Analysis of the thiol proteome showed few alterations to specific pathways mapped to the Kyoto Encyclopedia of Genes and Genomes Pathway database, but did reveal significant increases in concentrations of proteins associated with glycolysis/gluconeogenesis in the VYS and decreased concentrations proteins associated with ribosome biogenesis and function in the EMB. A subset of proteins elevated by >2-23-fold in the VYS were identified as serum (blood) proteins and represent the maternal-side proteins captured by the VYS and which are not degraded in the lysosomes as a result of leupeptin's inhibitory action. The observed constellation of proteins decreased in the EMB by leupeptin represent proteins from several adaptive pathways that are commonly altered in responses to amino acid starvation. These studies show clear differential responses to protease inhibition in VYS and EMB during organogenesis and suggest the possibility for additional roles of redox regulation, cellular adaptations and metabolic insufficiency caused by protease inhibition.
... A great deal of research has been focused on understanding the replication cycle of HIV, [31][32][33][34][35] in search of new drug targets and agents. [36][37][38][39][40][41] There is still a need for improvements in the selection of currently available anti-retroviral therapy: drugs of higher potency and those with better tolerability (to encourage adherence to the treatment regimen) and better resistance profiles are sought. HIV-1 life cycle. ...
... Many lines of evidence from biochemical and virological studies indicate that retroviral proteolysis is indeed tightly regulated, and that this regulation occurs through sequential cleavage at individual sites within the Gag polyprotein (Fig. 7). Amino acid sequences at the different PR recognition sites vary, and kinetic parameters determined in vitro using PR and synthetic peptides differ greatly between individual sites (reviewed in (Tozser and Oroszlan, 2003)). These differences are reflected in different cleavage rates at individual sites when Gag or Gag-Pro-Pol are used as substrates in vitro (Pettit et al., 2004(Pettit et al., , , 2002(Pettit et al., , , 2005(Pettit et al., , , 1994. ...
Article
Proteolytic processing of viral polyproteins is essential for retrovirus infectivity. Retroviral proteases (PR) become activated during or after assembly of the immature, non-infectious virion. They cleave viral polyproteins at specific sites, inducing major structural rearrangements termed maturation. Maturation converts retroviral enzymes into their functional form, transforms the immature shell into a metastable state primed for early replication events, and enhances viral entry competence. Not only cleavage at all PR recognition sites, but also an ordered sequence of cleavages is crucial. Proteolysis is tightly regulated, but the triggering mechanisms and kinetics and pathway of morphological transitions remain enigmatic. Here, we outline PR structures and substrate specificities focusing on HIV PR as a therapeutic target. We discuss design and clinical success of HIV PR inhibitors, as well as resistance development towards these drugs. Finally, we summarize data elucidating the role of proteolysis in maturation and highlight unsolved questions regarding retroviral maturation. Copyright © 2015 Elsevier Inc. All rights reserved.
... For all retroviruses, the completion of the viral budding process correlates with the activation of the viral protease by an unknown mechanism, and, as the structural (Gag) polyproteins are cleaved by the viral protease, maturation of the immature virus-like particle into an infectious virion [226]. It is generally believed that processing of precursor polyproteins is ordinarily delayed until after virus release. ...
Article
Full-text available
Viral proteases are critical enzymes for the maturation of many human pathogenic viruses and thus are key targets for direct acting antivirals (DAAs). The current viral pandemic caused by SARS-CoV-2 is in dire need of DAAs. The Main protease (Mpro) is the focus of extensive structure-based drug design efforts which are mostly covalent inhibitors targeting the catalytic cysteine. ML188 is a non-covalent inhibitor designed to target SARS-CoV-1 Mpro, and provides an initial scaffold for the creation of effective pan-coronavirus inhibitors. In the current study, we found that ML188 inhibits SARS-CoV-2 Mpro at 2.5 µM, which is more potent than against SAR-CoV-1 Mpro. We determined the crystal structure of ML188 in complex with SARS-CoV-2 Mpro to 2.39 Å resolution. Sharing 96% sequence identity, structural comparison of the two complexes only shows subtle differences. Non-covalent protease inhibitors complement the design of covalent inhibitors against SARS-CoV-2 main protease and are critical initial steps in the design of DAAs to treat CoVID 19.
Article
Processing of human immunodeficiency virus (HIV) proteins by the HIV-1 protease is essential for HIV infectivity. In addition, several studies have revealed cleavage of human proteins by this viral protease during infection; however, no large-scale HIV-1 protease degradomics study has yet been performed. To identify putative host substrates in an unbiased manner and on a proteome-wide scale, we used positional proteomics to identify peptides reporting protein processing by the HIV-1 protease, and a catalogue of over 120 cellular HIV-1 protease substrates processed in vitro was generated. This catalogue includes previously reported substrates as well as recently described interaction partners of HIV-1 proteins. Cleavage site alignments revealed a specificity profile in good correlation with previous studies, even though the ELLE consensus motif was not cleaved efficiently when incorporated into peptide substrates due to subsite cooperativity. Our results are further discussed in the context of HIV-1 infection and the complex substrate recognition by the viral protease.
Article
Full-text available
Retrovirus assembly and maturation involve folding and transport of viral proteins to the virus assembly site followed by subsequent proteolytic cleavage of the Gag polyprotein within the nascent virion. We report that inhibiting proteasomes severely decreases the budding, maturation, and infectivity of HIV. Although processing of the Env glycoproteins is not changed, proteasome inhibitors inhibit processing of Gag polyprotein by the viral protease without affecting the activity of the HIV-1 viral protease itself, as demonstrated by in vitro processing of HIV-1 Gag polyprotein Pr55. Furthermore, this effect occurs independently of the virus release function of the HIV-1 accessory protein Vpu and is not limited to HIV-1, as proteasome inhibitors also reduce virus release and Gag processing of HIV-2. Electron microscopy analysis revealed ultrastructural changes in budding virions similar to mutants in the late assembly domain of p6gag, a C-terminal domain of Pr55 required for efficient virus maturation and release. Proteasome inhibition reduced the level of free ubiquitin in HIV-1-infected cells and prevented monoubiquitination of p6gag. Consistent with this, viruses with mutations in PR or p6gag were resistant to detrimental effects mediated by proteasome inhibitors. These results indicate the requirement for an active proteasome/ubiquitin system in release and maturation of infectious HIV particles and provide a potential pharmaceutical strategy for interfering with retrovirus replication.
Article
Full-text available
Host proteins are incorporated into retroviral virions during assembly and budding. We have examined three retroviruses, human immunodeficiency virus type 1 (HIV-1), simian immunodeficiency virus (SIV), and Moloney murine leukemia virus (Mo-MuLV), for the presence of ubiquitin inside each of these virions. After a protease treatment to remove exterior viral as well as contaminating cellular proteins, the proteins remaining inside the virion were analyzed. The results presented here show that all three virions incorporate ubiquitin molecules at approximately 10% of the level of Gag found in virions. In addition to free ubiquitin, covalent ubiquitin-Gag complexes were detected, isolated, and characterized from all three viruses. Our immunoblot and protein sequencing results on treated virions showed that approximately 2% of either HIV-1 or SIV p6Gag was covalently attached to a single ubiquitin molecule inside the respective virions and that approximately 2 to 5% of the p12Gag in Mo-MuLV virions was monoubiquitinated. These results show that ubiquitination of Gag is conserved among these retroviruses and occurs in the p6Gag portion of the Gag polyprotein, a region that is likely to be involved in assembly and budding.
Article
Full-text available
We have investigated the cellular uptake of Gag p24 shortly after exposure of cells to human immunodeficiency virus (HIV) particles. In the absence of envelope glycoprotein on virions or of viral receptors or coreceptors at the cell surface, p24 was incorporated in intracellular vesicles but not detected in the cytosolic subcellular fraction. When appropriate envelope-receptor interactions could occur, the nonspecific vesicular uptake was still intense and cytosolic p24 represented 10 to 40% of total intracellular p24. The measurement of cytosolic p24 early after exposure to HIV type 1 is a reliable assay for investigating virus entry and early events leading to authentic cell infection.
Article
Full-text available
We have evaluated a possible role for human immunodeficiency virus type 1 protease during early steps of replication. For these studies, a specific inhibitor of human immunodeficiency virus protease, Ro31-8959, was used. Synthesis of viral cDNA, its integration into cellular DNA, and its transcription were determined during a one-step, acute infection of MT-4 cells. No consistent difference in any of these parameters was noted between control-infected cultures and those treated with protease inhibitor. However, no infectious progeny virus was produced in treated cultures, and thus spread of infection was severely restricted. Our results do not support an essential activity of viral protease in early steps of replication but are in line with its established role in gag and gag-pol processing and in maturation to infectious progeny virus.
Article
We asked whether human immunodeficiency virus type 1 (HIV-1) protease plays a major role in the early stages of infection (i.e. from viral entry to reverse transcription) by using various protease inhibitors (saquinavir, ritonavir, and KNI-272). When assessed in the two-day multinuclear activation of a galactosidase indicator (MAGI) assay, involving a single cycle of HIV-1 replication, all protease inhibitors failed to block infection of HeLa-CD4-LTR-β-gal cells by HIV-1, while reverse transcriptase (RT) inhibitors (AZT and ddI) completely blocked the infection. Moreover, when HIV-1 proviral DNA synthesis was examined by polymerase chain reaction in HeLa-CD4-LTR-β-gal cells exposed to HIV-1 and cultured in the presence of protease inhibitors, a significant amount of proviral DNA was detected, while no proviral DNA synthesis was detected when the cells were cultured in the presence of RT inhibitors. Protease inhibitors also failed to block chloramphenicol acetyltransferase (CAT) expression in HLCD4-CAT cells exposed to HIV-1, while RT inhibitors completely suppressed CAT expression. These results strongly suggest, contrary to a previous report by Nagy et al. (1994), that HIV-1 protease does not play a major role in the early stages of infection.
Article
Host proteins are incorporated both on and inside human immunodeficiency virus type 1 (HIV-1) virions. To identify cellular proteins inside HIV-1, virion preparations were treated by a protease-digestion technique that removes external host proteins, allowing for the study of the proteins inside the virus. Treated HIV-1 preparations were analyzed by immunoblot, high-pressure liquid chromatography, and protein sequence analyses. These analyses identified several cellular proteins inside HIV-1: elongation factor 1alpha, glyceraldehyde-3-phosphate dehydrogenase, HS-1, phosphatidylethanolamine-binding protein, Pin1, Lck, Nm23-H1, and the C-terminal tail of CD43. Several of these proteins were found as fragments of their full-sized proteins that appear to be generated by our protease treatment of the virions, the HIV-1 protease, or a cellular protease. Recent advances in cell biology and biochemistry have identified some of these proteins as actin-binding proteins. These results support the hypothesis that actin filaments are incorporated into the virion and may provide additional clues for the understanding of the interaction between viral and cellular proteins during assembly and budding.
Article
The amino- and carboxyl-terminal amino acid sequences of proteins (p10, p12, p15, and p30) coded by the gag gene of Rauscher and AKR murine leukemia viruses were determined. Among these proteins, p15 from both viruses appears to have a blocked amino end. Proline was found to be the common NH(2) terminus of both p30s and both p12s, and alanine of both p10s. The amino-terminal sequences of p30s are identical, as are those of p10s, while the p12 sequences are clearly distinctive but also show substantial homology. The carboxyl-terminal amino acids of both viral p30s and p12s are leucine and phenylalanine, respectively. Rauscher leukemia virus p15 has tyrosine as the carboxyl terminus while AKR virus p15 has phenylalanine in this position. The compositional and sequence data provide definite chemical criteria for the identification of analogous gag gene products and for the comparison of viral proteins isolated in different laboratories. On the basis of amino acid sequences and the previously proposed H-p15-p12-p30-p10-COOH peptide sequence in the precursor polyprotein, a model for cleavage sites involved in the post-translational processing of the precursor coded for by the gag gene is proposed.
Article
The envelope glycoprotein of human immunodeficiency virus (HIV) initiates infection by mediating fusion of the viral envelope with the cell membrane. Fusion activity requires proteolytic cleavage of the gp160 protein into gp120 and gp41 at a site containing several arginine and lysine residues. Activation at basic cleavage sites is observed with many membrane proteins of cellular and viral origin. We have recently found that the enzyme activating the haemagglutinin of fowl plague virus (FPV), an avian influenza virus, is furin. Furin, a subtilisin-like eukaryotic endoprotease, has a substrate specificity for the consensus amino-acid sequence Arg-X-Lys/Arg-Arg at the cleavage site. We show here that the glycoprotein of HIV-1, which has the same protease recognition motif as the FPV haemagglutinin, is also activated by furin.
Article
The processing of the human immunodeficiency virus (HIV) gag and gag-pol precursor proteins by the virus-encoded protease is an essential step in maturation of infectious virus particles. Like most retroviral proteases, the HIV protease belongs to the aspartyl-protease family and can be inhibited by specific inhibitors. Twenty-four synthetic peptides known to be inhibitors of human renin were tested for inhibition of HIV replication in tissue cultures. One of them, a synthetic peptide analogue, SR41476, which has been shown to be a specific inhibitor of purified recombinant HIV1 protease in vitro, totally blocked infection with different isolates including the HIV1 LAV prototype, the highly cytopathic Zairian isolate HIV1 NDK, and HIV2 ROD, both in primary blood lymphocytes (PBL) and in the lymphoid cell lines MT4 and CEM, for at least 3 weeks. It also significantly reduced virus replication in chronically infected CEM cells, without any effect on cell proliferation. Radioimmunoprecipitation assay revealed that the inhibitor blocked processing of polyprotein precursors p55 gag and p40 gag into a mature form of gag proteins, p25 and p18. Synthetic peptide analogue SR 41476, when added before infection, efficiently inhibited formation of HIV DNA provirus and successfully suppressed synthesis of HIV-specific proteins. These results imply that the HIV protease inhibitor not only inhibited virus maturation in the late phase of the HIV replication cycle, but also interfered in the early phase, before the provirus was formed. This mechanism of antiviral activity provides new possibilities and strategies for AIDS chemotherapy.