ArticlePDF Available

HIV-1 protease processes procaspase 8 to cause mitochondrial release of cytochrome c, caspase cleavage and nuclear fragmentation

Authors:

Abstract and Figures

Infection of T cells with HIV-1 induces apoptosis and modulates apoptosis regulatory molecules. Similar effects occur following treatment of cells with individual HIV-1 encoded proteins. While HIV-1 protease is known to be cytotoxic, little is known of its effect on apoptosis and apoptosis regulatory molecules. The ability of HIV-1 protease to kill cells, coupled with the degenerate substrate specificity of HIV-1 protease, suggests that HIV-1 protease may activate cellular factor(s) which, in turn, induce apoptosis. We demonstrate that HIV-1 protease directly cleaves and activates procaspase 8 in T cells which is associated with cleavage of BID, mitochondrial release of cytochrome c, activation of the downstream caspases 9 and 3, cleavage of DFF and PARP and, eventually, to nuclear condensation and DNA fragmentation that are characteristic of apoptosis. The effect of HIV-1 protease is not seen in T cell extracts which have undetectable levels of procaspase 8, indicating a specificity and requirement for procaspase 8.
Content may be subject to copyright.
HIV-1 protease processes procaspase 8 to cause
mitochondrial release of cytochrome
c
, caspase cleavage
and nuclear fragmentation
Z Nie
1,2
, BN Phenix
1,2
, JJ Lum
1,2
, A Alam
3
, DH Lynch
4
,
B Beckett
2
, PH Krammer
5
, RP Sekaly
3
and AD Badley*
,1,2,6
1
Department of Biochemistry, Microbiology and Immunology, University of
Ottawa, Ottawa, Ontario, Canada
2
Ottawa Health Research Institute, Ottawa, Ontario, Canada
3
Institut de Recherches Cliniques de Montreal, Montreal, Quebec, Canada
4
Immunex Corporation, Seattle, Washington, USA
5
Tumor Immunology Program, German Cancer Research Center, Heidelberg,
Germany
6
Division of Infectious Diseases, Mayo Clinic and Foundation, Rochester,
Minnesota, USA
* Corresponding author: Andrew Badley, MD, FRCPC, Division of Infectious
Diseases, Mayo Clinic, 200 First Street NW, Rochester, Minnesota 55905,
USA. Tel: 507-255-7761; Fax: 507-255-7767.
Received 15.3.02; revised 17.6.02; accepted 17.6.02
Edited by RA Knight
Abstract
Infection of T cells with HIV-1 induces apoptosis and
modulates apoptosis regulatory molecules. Similar effects
occur following treatment of cells with individual HIV-1
encoded proteins. While HIV-1 protease is known to be
cytotoxic, little is known of its effect on apoptosis and
apoptosis regulatory molecules. The ability of HIV-1 protease
to kill cells, coupled with the degenerate substrate specificity
of HIV-1 protease, suggests that HIV-1 protease may activate
cellular factor(s) which, in turn, induce apoptosis. We
demonstrate that HIV-1 protease directly cleaves and
activates procaspase 8 in T cells which is associated with
cleavage of BID, mitochondrial release of cytochrome
c
,
activation of the downstream caspases 9 and 3, cleavage of
DFF and PARP and, eventually, to nuclear condensation and
DNA fragmentation that are characteristic of apoptosis. The
effect of HIV-1 protease is not se en in T cell extracts which ha ve
undetectable levels of procaspase 8, indicating a specificity
and requirement for procaspase 8.
Cell Death and Differentiation (2002) 9, 1172 ± 1184.
doi:10.1038/sj.cdd.4401094
Keywords: apoptosis; HIV; HIV-1 protease; caspase 8; mitochon-
drial permeability transition pore complex, cytochrome c
Abbreviations: AICD, activation induced cell death; ATCC,
American type cell culture; ATP, adenine trinucleotide phosphate;
BA, bongkrekic acid; BSA, bovine serum albumin; CHAPS,
cholamidopropyl dimethylammonio propane sulfonate; DFF, DNA
fragmentation factor; DMSO, dimethyl sulfoxide; DTT, dithiothreitol;
EDTA, ethylene diamine tetracetic acid; EGTA, ethylene glycol
tetracetic acid; FLIP, FLICE-like inhibitory peptide; HEPES,
hydroxyethyl piperazine ethane sulfonic acid; HIV, human
immunode®ciency virus; HIV-1 PI, HIV-1 protease inhibitor; HPLC,
high performance liquid chromatography; HRP, horseradish
peroxidase; PAGE, poly crylamide gel electropheresis; PARP, poly
(ADP Ribose) polymerase; PBL, peripheral blood lymphocyte;
PMSF, phenylmethylsulfonyl ¯uoride; SDS, sodium dodecyl
sulphate; TRAIL, TNF related apoptosis inducing ligand
Introduction
HIV-1 infection results in CD4 T cell apoptosis which
contributes to CD4 T cell depletion in infected individuals.
Multiple mechanisms have been proposed to explain
enhanced CD4 T cell apoptosis in HIV-1 infected persons.
HIV-1 infected accessory cells, including macrophages,
develop the ability to induce apoptosis of autologous
uninfected CD4 T cells by producing the apoptosis inducing
ligand, Fas (APO-1/CD95) Ligand (FasL).
1±10
AICD of T cells
is a physiologic response to activation
11 ± 14
which is greater in
HIV-1 infected individuals than in uninfected controls,
15 ± 19
and is potentially induced by tat and/or gp120 cross linking the
CD4 receptor
3,20 ± 22
resulting in increased expression of Fas
Ligand, TNF or TRAIL.
17,23
A third form of HIV-1 induced CD4
T cell death follows direct infection of a CD4 T cell with HIV-1,
and is independent of Fas receptor ligation.
8,15,24,25
While numerous HIV-1 proteins, including tat,
26 ± 28
gp120,
20±22
Nef,
29±32
vpr,
33 ± 35
and protease
36 ± 39
have been
implicated as direct mediators of infected CD4 T cell death, the
molecular mechanisms , whereby some of these HIV-1 specific
proteins induce apoptosis, including the mechanisms asso-
ciated with HIV-1 protease induced death, are unclear.
HIV-1 protease, a late regulatory protein in the HIV-1 life
cycle, functions as a homodimer
40
to cleave HIV-1
polyprotein. While ectopic expression of HIV-1 protease
induces apoptosis in a variety of cell types, including
human CD4 T cells,
36±39
coincubation of nuclei with HIV-1
protease does not induce the nuclear changes of
apoptosis,
40
suggesting that cytosolic factor(s) must be
activated by HIV-1 protease which in turn either directly or
indirectly causes nuclear fragmentation. The presence of
active HIV-1 protease within the cytosolic fraction of
infected cells
41,42
raises the possibility that cleavage of
non viral proteins by HIV-1 protease may contribute to the
cytotoxicity of HIV-1 infection. In support of this view, HIV-1
protease substrate specificity is not restricted to viral
proteins, since Bcl-2, actin, laminin B and pro-interleukin-
1 are cleaved by HIV-1 protease both in vitro and in
vivo.
36,43
Although some of the proteins cleaved by HIV-1
protease
36,43
are important in the regulation of apoptosis,
none alone is sufficient to induce apoptosis. We propose
Cell Death and Differentiation (2002) 9, 1172 ± 1184
ã
2002 Nature Publishing Group All rights reserved 1350-9047/02 $25.00
www.nature.com/cdd
that HIV-1 protease cleaves alternate apoptosis regulatory
molecules in such a manner that they develop the ability to
induce apoptosis.
Results
HIV-1 protease induces HeLa nuclear apoptosis
and DNA fragmentation in cell-free system
To determine if HIV-1 protease induces nuclear fragmenta-
tion, we modified a previously described cell-free system.
44,45
Cytoplasmic extracts from Jurkat T cells were treated with or
without HIV-1 protease and co-incubated with HeLa nuclei.
The nuclear membranes and chromatin of nuclei incubated
with untreated cytoplasmic extracts were intact (Figure 1A), in
contrast to nuclei coincubated with HIV-1 protease treated
cytoplasmic extracts which were marginated (Figure 1B) and/
or fragmented (Figure 1C). These nuclear effects of HIV-1
protease were inhibited by an HIV-1-PI (Figure 1D). Similarly,
nuclei incubated with HIV-1 protease treated cytoplasmic
extracts developed internucleosomal DNA cleavage (as
determined by DNA ladder analysis) which was also inhibited
by HIV-1-PI (Figure 2A). As a control, the human aspartyl
protease renin was used to treat cytoplasmic extracts, and, by
contrast, the renin treated cytoplasmic extracts did not induce
DNA laddering (Figure 2B), despite maintaining activity as
determined by cleavage of the fluorogenic renin substrate 1
(fluorescence of control cytosols=0 relative fluorescence
units, fluorescence of cytosols=22431 relative fluorescence
units). Since HIV-1 protease alone does not directly induce
the nuclear changes of apoptosis
40
(data not shown),
cytoplasmic signals must necessarily be activated by HIV-1
protease which, in turn, leads to the nuclear events of
apoptosis.
Caspase cascade is activated in cell extracts
treated with HIV-1 protease
We next assessed procaspase 8 and procaspase 3
processing after treatment of Jurkat cytoplasmic extracts with
HIV-1 protease. Both the 18 kd active fragment of caspase 8
and the 17 kd active fragment from caspase 3
46,47
were
detected following HIV-1 protease treatment but not in control
cytosols nor in renin treated cytoplasmic extracts (Figure 3A).
In HIV-1 protease treated, but not untreated cytoplasmic
extracts, cytochrome cwas released from mitochondria into
the cytoplasmic compartment (Figure 3B) in a comparable
manner to the release of cytochrome cseen with
recombinant active caspase 8 or Granzyme B, indicating
mitochondrial activation in treated cytoplasmic extracts.
Figure 1 HIV-1 protease induces the nuclear changes of apoptosis. Jurkat cytosols (1 mg) were treated with recombinant HIV-1 protease at 308C for 3 h and then
co-incubated with HeLa nuclei. Treated nuclei were imaged under microscopy by Hoechst 33342 staining. (A) Nuclei incubated with Jurkat cytosols without HIV-1
protease treatment. (B) Nuclei incubated with Jurkat cytosols treated with HIV-1 protease, resulting in fragmentation of the nuclear membrane and chromatin
condensation or (C) margination of chromatin. (D) The induction of apoptotic changes were completely inhibited by HIV-1-PI
HIV-1 protease activates caspase 8
ZNieet al
1173
Cell Death and Differentiation
Following the mitochondrial release of cytochrome c
48,49
into cytosols, cytosolic cytochrome ccomplexes with
APAF-1 in the presence of dATP to form the apoptosome
which allows the autoactivation of procaspase 9.
50,51
In
those samples where cytochrome crelease was seen,
procaspase 9 cleavage was also present, suggesting
formation of the apoptosome and downstream caspase
activation (Figure 3C).
We next determined whether HIV-1 protease mediated
cleavage of procaspase 8 is responsible for mitochondrial
activation. BID is a cytosolic member of the Bcl-2 family of
apoptosis regulatory proteins
52
that is cleaved by caspase
8 to create a truncated form of BID (tBID) which
translocates to mitochondria and causes the release of
cytochrome cinto the cytosol.
53
The p15 tBID form was
detected in the HIV-1 protease treated cytoplasmic extracts
but not in untreated cytosols (Figure 3D). Conversely, while
HIV-1 protease may cleave Bcl-2
43
we did not detect Bcl-2
cleavage in this assay (Figure 3D) although it was
observed after 4 h (data not shown). Following mitochon-
drial activation and downstream effector caspase activation,
cellular substrates, including PARP, are cleaved. Consis-
tent with our data indicating caspase activation in HIV-1
protease treated cytoplasmic extracts, but not untreated
cytoplasmic extracts, the 85 kd fragment of activated PARP
was seen only in HIV-1 protease treated cytoplasmic
extracts (Figure 3D). These data suggest that activated
caspase 8 cleaves BID to initiate the mitochondrial events
which lead to apoptosis. Kinetic analysis of cleavage of
procaspase 8 and cytochrome crelease was performed at
308C to slow the reaction, and analysed using Western
blot. In these experiments cytochrome cwas released after
the cleavage of procaspase 8 into its 18 kd active
fragments (Figure 3E).
Activation of caspase 8 leads to the activation of
downstream caspases
We next determined the kinetics of caspase activation. HIV-1
protease induced the processing of procaspase 8 as early as
1 min after adding HIV-1 protease at 378C (Figure 4A), and
cleavage of procaspase 3 into its 17 kd active fragment was
seen within 5 m. The relationship between caspase 8
cleavage and the cleavage of caspase 3, 9 and DFF were
next evaluated in reaction mixtures incubated at 308C. In
these experiments HIV-1 cleavage of both procaspase 8 and
3 induced by HIV-1 protease was inhibited by HIV-1-PI pre-
treatment, but only procaspase 3 cleavage was inhibited by
the caspase 8 inhibitor (IETD-fmk) (Figure 4B). The lack of
procaspase 8 inhibition by z-IETD-fmk indicates that
procaspase 8 activation is a consequence of HIV-1 protease,
rather than a result of autocatalysis. Thus both the timing of
caspase 3 cleavage (Figure 4A) and its inhibition by z-IETD-
fmk (Figure 4B) indicate that the cleavage of procaspase 3
depends upon prior caspase 8 activation. Furthermore,
cleavage of procaspase 9 occurred after the cleavage of
procaspase 8 (Figure 4B) and was inhibited by saquinavir and
partially inhibited by z-IETD-fmk (Figure 4B). Therefore both
caspase 3 and 9 activation occur after caspase 8 activation.
We also determined that cleavage of DFF (a DNAse,
activated by caspase 3, that contributes to nuclear fragmenta-
tion) into its 10 kd form occurred after 4 h in the treated
cytosols, and its cleavage was inhibited by HIV-1-PI and by z-
IETD-fmk (Figure 4B). These results demonstrate that HIV-1
protease treatment of cytoplasmic extracts results in
procaspase 8 processing which precedes and contributes to
processing of caspases 9 and 3 as well as DFF.
HIV-1 protease cleaves caspase 8 but not
caspase 3
The ability of HIV-1 protease to cleave pro-interleukin 1 into its
active subunits
54
infers that it may function as a caspase, a
suggestion that is supported by our data in Jurkat cytoplasmic
A
B
Figure 2 HIV-1 protease induces internucleosomal DNA fragmentation. (A)
DNA gel from nuclei incubated with Jurkat cytosols in the presence or absence
of HIV-1 protease with or without HIV-1 PI. (B) DNA gel from nuclei incubated
with Jurkat cytosols and treated with or without HIV-1 protease or renin
(control)
Cell Death and Differ entiati on
HIV-1 protease activates caspase 8
ZNieet al
1174
extracts showing that HIV-1 protease cleaves and activates
procaspase 8. To investigate this possibility further, recombi-
nant GST-caspase 8 was directly incubated with HIV-1
protease (Figure 5A). Within 1 min of co-incubation of
recombinant GST-procaspase 8 with HIV-1 protease, cas-
pase 8 is cleaved specifically by HIV-1 protease, as
demonstrated by the lack of autocatalysis of GST-caspase 8
and the inhibition of HIV-1 protease cleavage by HIV-1-PI
(HIV-1-PI does not inhibit caspase 8 activity (data not
shown)). Importantly, coincubation of HIV-1 protease with
full-length recombinant GST-caspase 8 generates p18
fragments, which have previously been associated with
caspase 8 activity.
47,55,56
To confirm the activity of the p18
caspase 8 fragments, we tested the ability of GST-caspase 8
treated with HIV-1 protease to cleave caspase 3, yet such
experiments did not result in caspase 3 cleavage (data not
shown). However, when HIV-1 protease was added after
GST-caspase 8 was cleaved by HIV-1 protease (to inhibit
remaining HIV-1 protease activity) (Figure 5B, top), and then
cytoplasmic extracts added, caspase 3 was cleaved (Figure
5B, bottom), suggesting the requirement of a mitochondrial
amplification step to cleave caspase 3. In these experiments
the effects of GST-caspase 8 cleavage products on caspase
3 were inhibited by z-IETD-fmk (Figure 5B, bottom). In
contrast to our results with GST-caspase 8, incubation of
recombinant caspase 3 with HIV-1 protease did not result in
cleavage, yet co-incubation of caspase 3 with Granzyme B
did, as previously described
57
(Figure 5C).
HIV-1 protease cleavage of procaspase 8 occurs at
an atypical site
The pattern of procaspase 8 cleavage that follows HIV-1
protease cleavage appears distinct from that seen with active
caspase 8 treatment (compare Figures 5A and B with Figures
3A, E and 4A, B), suggesting that the HIV-1 protease
cleavage site is different than the usual caspase 8 cleavage
site. We instead propose that HIV-1 protease generates
active caspase 8 (cleaved at an atypical site, Figure 5A, B),
which then activates more procaspase 8 (cleaved at the
typical site) resulting in the generation of p43, p41 and p18
fragments (Figures 3A, E and 4A, B).
To assess this possibility further, two sets of experi-
ments were performed. First we mutated the typical
cleavage of caspase 8. The initial cleavage event of
procaspase 8 activation occurs at ASP374,
56
within the
domain VETDSEEQ. Using a sequence coupled predictive
method of Markov chain theory,
58
this sequence would be
predicted to be cleaved by HIV-1 protease with a high
degree of likelihood. We therefore mutated this domain to
VDPDSDKQ, using site directed mutagenesis, as this
sequence is extremely unlikely to be cleaved by HIV-1
protease.
58
Both WT and mutant forms of GST-procaspase
8 were then reacted with HIV-1 protease. Analysis of
cleavage products by Western blot revealed identical
banding patterns, suggesting that HIV-1 protease cleavage
of procaspase 8 does not occur at this site.
To further address whether HIV-1 protease initiates
cleavage at this site, HIV-1 protease was incubated with
fluorogenic substrate z-IETD-AFC (Figure 6). Both active
caspase 8 and Granzyme B caused cleavage of z-IETD-
AFC, yet consistent with our mutational data, HIV-1
protease did not directly cleave z-IETD-AFC, supporting
the concept that HIV-1 protease activates procaspase 8 at
a site distinct from the typical activation site.
HIV-1 protease induced apoptotic signaling
requires procaspase 8
Our cumulative data thus demonstrate a direct effect of HIV-1
protease on procaspase 8, which is associated with the
downstream events of apoptosis including cleavage of BID,
release of cytochrome c, activation of caspases 9 and 3, as
well as cleavage of DFF and PARP. It remains possible that
HIV-1 protease initiated cleavage of other factors (e.g. other
initiator caspases) may also occur to initiate apoptotic
signaling. Thus, we assessed the ability of HIV-1 protease
to initiate apoptotic signaling in cells which are deficient in
procaspase 8. Cytosolic extracts of JB6 cells and I9.2 cells
which are a procaspase 8 deficient T cell derivatives were
treated with or without HIV-1 protease and cleavage of
procaspase 8, BID, procaspase 3 assessed. As expected,
while JB6 and I9.2 cells had undetectable levels of
procaspase 8, Jurkat T cell procaspase 8 was processed by
HIV-1 protease. Only in the Jurkat T cell extracts treated with
HIV-1 protease, was there any evidence of cleavage of BID or
of procaspase 3 (Figure 7), indicating that the presence of
procaspase 8 in Jurkat T cells is required for activation of the
downstream apoptotic signaling events, since the absence of
procaspase 8 in JB6 and I9.2 cells prevents downstream
apoptotic signaling.
Direct infection of HIV-1 causing cell death is
correlated with HIV protease expression and
requires active caspase 8
To determine whether HIV-1 protease expression is corre-
lated with the induction of apoptosis, we analyzed the
expression of protease in relation to the timing of apoptosis
in Jurkat T cells acutely infected with HIV-1. In this model of
acute HIV-1 infection, cell death by apoptosis occurs several
days following infection,
15
is inhibited by z-VAD-fmk, z-IETD-
fmk (Figure 8A) and by Saquinavir (data not shown), and is
associated with caspase 8 and PARP cleavage (Figure 8B).
Further cell death coincides with detectable expression of
HIV-1 protease (Figure 8C). Freshly isolated PBL were also
collected from six HIV-1 negative controls and from two
untreated patients infected with HIV-1 were analyzed for
expression of HIV-1 protease and for PARP cleavage into an
85 kd apoptosis characteristic fragments.
59,60
In control
patient 1 (who had an upper respiratory tract infection) and
HIV-1 patients 2 to 6 the 85 kd PARP fragment was present,
indicating that PBLs from these patients were undergoing
apoptosis (Figure 8D). Conversely the 85 kd PARP was not
present in control patient 2 and HIV patients 1. Expression of
HIV-1 protease was seen only in HIV-1 patients 2 ± 6 who had
high levels of viral replication (4500,000 copies/ml) and
importantly HIV-1 protease was not detected in HIV-1 patient
1 who had a low level of viral replication (1600 copies/ml).
Thus, given previous literature which demonstrate that HIV-1
Cell Death and Differentiation
HIV-1 protease activates caspase 8
ZNieet al
1175
A
B
C
Cell Death and Differ entiati on
HIV-1 protease activates caspase 8
ZNieet al
1176
protease cleaves actin into an HIV-1 protease specific pattern
in vivo, as well as in vitro,
36
our observations that apoptosis in
HIV-1 infection coincides with HIV-1 protease expression
support a possible role for HIV-1 protease apoptosis
associated with directly infected cells.
Discussion
The demonstration of caspase activation by HIV-1 protease is
significant for several reasons. First, the ability of HIV-1
protease to induce T cell apoptosis represents another
potential mechanism whereby HIV-1 may cause death of
HIV-1 infected T cells. This mechanism applies only to cells
directly infected by HIV-1, as addition of HIV-1 protease to cell
cultures does not influence cell viability (data not shown). The
relative importance of this mechanism, in comparison to other
proposed mechanisms of HIV-1 associated T cell depletion
(reviewed in
61
), including AICD, autologous cell mediated
killing, and direct virus induced killing associated with gp120,
Nef, Tat and/or Vr is however unclear. Secondly, the ability of
viruses to influence apoptosis has been well characterized,
62
and a number of virally encoded proteins have been shown to
interact with members of the caspase family to inhibit
apoptosis: these include baculovirus IAP and p35, Adeno-
virus E1B-19k, Cowpox Crm-A, Epstein Barr virus BHRF1,
62
and gamma herpes virus FLIP.
63
In contrast, HIV-1 protease
is an example of a virally encoded protein that activates
caspase 8 to promote apoptosis.
D
E
Figure 3 Caspase activation and mitochondrial release of cytochrome coccurs in Jurkat cytosols treated with HIV-1 protease. Jurkat cytosols were treated with
HIV-1 protease and cleavage of procaspases 8, 3, 9 and the caspase substrates, BID, Bcl-2 and PARP were assessed along with mitochondrial release of
cytochrome c.(A) Cleavage profiles of procaspase 8 and 3 indicating active p18 and p17 fragments respectively in the cytosols treated with HIV-1 protease, but not
those treated with renin. (B) Jurkat cytosols were incubated with HIV-1 protease, recombinant active caspase 8 or Granzyme B, fractionated and analysed for
cytochrome ccontent in the total reaction mixture, mitochondrial fractions or mitochondria free cytosolic fraction. (C) Cleavage profile of procaspase 9 indicating
p35 fragment in cytosols treated with HIV-1 protease, as well as cytochrome crelease from mitochondria. HSP 70 is analysed as a control mitochondria specific
protein. (D) The cleavage of BID, Bcl-2 and PARP induced by HIV-1 protease treatment of cytosols. PCNA is included as an internal control. (E) Jurkat cytosols
(1 mg) were treated with HIV-1 protease at 308C, and assayed at the indicated times for analysis of caspase 8 cleavage and cytochrome c release. PCNA was used
as an internal control
Cell Death and Differentiation
HIV-1 protease activates caspase 8
ZNieet al
1177
The HIV-1 genome is translated as polyprotein fusions
that require processing by HIV-1 protease. These poly-
proteins are processed by HIV-1 protease in two cellular
compartments: first, as membrane-associated polyproteins
that are cleaved for viral assembly and maturation, and
second, as free polyproteins within the cytosols
41,42
of
infected cells. Previous studies have shown that HIV-1
protease can induce apoptosis in both transfected and
microinjected cells.
43,64,65
Furthermore, a variety of cellular
proteins, including the antiapoptotic regulating protein Bcl-2
and cell structure proteins such as laminin B and
cytoskeleton proteins, are substrates of HIV-1 protease in
vitro and in vivo.
39,43,54,65,66
These observations suggest
that the degenerate substrate specificity of HIV-1 protease
allows protease to activate proteins which initiate apoptosis
cascades.
In the present study, we have developed a cell-free
system to characterize the mechanisms by which HIV-1
B
A
Figure 4 Activation of procaspase 8 by HIV-1 protease leads to cleavage of
downstream caspases. (A) At the indicated times, 100 mg cytosol proteins
were probed with anti-caspase 8 and 3. (B) In parallel cleavage of caspase 9
and DFF were assessed. As indicated either the caspase 8 inhibitor z-IETD-
fmk or HIV-1-PI were used
A
B
C
Figure 5 HIV-1 protease directly cleaves GST-procaspase 8 but not
procaspase 3. Purified recombinant GST-procaspase 8 (A) was incubated
for the indicated times with HIV-1 protease, with or without HIV-1-PI and
analyzed for cleavage. Reactions were stopped at the indicated times by
addition of gel loading buffer. (B) GST-caspase 8 was incubated with HIV-1
protease for 30 min and analyzed for caspase 8 cleavage (top). Thereafter
reactions were stopped by the addition of HIV-1 PI, and Jurkat cytosols added
and analyzed for caspase 3 cleavage (bottom). (C) Treatment of procaspase 3
with HIV-1 protease does not alter procaspase 3, whereas Granzyme B results
in cleavage of procaspase 3
Cell Death and Differ entiati on
HIV-1 protease activates caspase 8
ZNieet al
1178
protease induces apoptosis. Cell-free systems have been
successfully used to identify the apoptotic molecules and
their signal pathways.
44,45,67
In our system, treatment of
cytosols with HIV-1 protease initiates a pathway that
involves activation of both caspases and mitochondrial
events involved in apoptosis (Figure 9). Further, we
demonstrate that the apical and requisite event in this
pathway is the cleavage of procaspase 8 by HIV-1
protease, which in turn activates BID, causes mitochondrial
release of cytochrome c, activation of caspases 9 and 3 as
well as cleavage of DFF and PARP. The requirement for
mitochondria in this apoptosis cascade is demonstrated by
observations that GST caspase 8 activated by HIV-1
protease does not cleave caspase 3. Only when GST
caspase 8 was incubated with HIV-1-PR, PI added (to
inhibit protease) and the entire reaction added to
cytoplasmic extract was caspase 3 activated (Figure 5B).
However, as it is now recognized that activated caspase 8
can initiate apoptosis directly via caspase 3 (type 1
pathway) or indirectly via mitochondrial activation, cyto-
chrome-crelease and caspase 9 processing (type 2
pathway),
68,69
we cannot exclude the possibility that HIV-
1 protease mediated apoptosis may involve both type 1 as
well as type 2 signaling pathways. Indeed, when T cell
extracts treated with HIV-1 protease in the presence or
absence of the mitochondrial PTPC inhibitor BA were
analysed for caspase 3 and caspase 9 activation, BA
resulted in partial, but incomplete inhibition of caspase 3
and 9 activation, thereby indicating that both type I and type
II pathways are likely involved (data not shown). The
results are consistent with previous work which demon-
strate that activation of procaspase 8 is sufficient to induce
changes in a cell-free system that are similar to those seen
during apoptosis in vivo.
47
In the present study we demonstrate that procaspase 8
is required for HIV-1 protease induced apoptosis, as both
JB6 and I9.2 cells which are deficient in procaspase 8 do
not develop the molecular changes of apoptosis following
HIV-1 protease treatment. However, since our evidence
that HIV protease activates caspase 8 physiologically is
indirect, it remains possible that it may also act on different
substrates to initiate death pathways. Additional studies are
underway to address these possibilities.
Treatment of HIV-1 infected patients with inhibitors of
HIV-1 protease has dramatically reduced both morbidity
and mortality associated with this infection. Thus far, two
reasons for the improved outcomes are apparent: first,
protease inhibitors are potent inhibitors of viral replication
70
and second, this class of drugs possesses intrinsic
immunomodulatory properties including antiapoptotic ef-
fects.
61,71
We suggest that direct inhibition of HIV-1
protease also reduces protease induced apoptosis of
infected cells to further reduce HIV-1 associated T cell
death. Further research is therefore required to determine
the contribution of this form of cell death on the
pathogenesis of HIV-1 disease, and the effect of HIV-1
protease mutations on the pathogenesis of HIV-1 induced
immunodeficiency.
Materials and Methods
Preparation of cell-free extracts
Cell-free extracts were freshly prepared from human Jurkat T
lymphoblastoid cells (ATCC, Rockville, MD, USA) as described
previously
44,45
with some modifications. Briefly, cells (0.5610
6
cells/ml) were harvested by centrifugation at 16006gfor 5 min at
48C. The cell pellet was washed twice with ice-cold PBS (pH 7.4),
followed by a single wash with ice-cold caspase buffer (20 mM PIPES,
100 mM NaCl, 10 mM DTT, 1 mM EDTA, 0.1% CHAPS, 250 mM
sucrose, pH 7.2).
72
After centrifugation, the cells were resuspended
with two volumes of ice-cold complete caspase buffer which was
supplemented with protease inhibitors (100 mmPMSF,10mg/ml
leupeptin, 2 mg/ml aprotinin) and then transferred to a 2-ml dounce
homogenizer. After sitting on ice for 15 min, the cells were disrupted
with 50 strokes of B-type pestle (Fisher Scientific Ltd, Nepean, ON,
Canada). Cell disruption (495%) was confirmed by examination of
5ml aliquot of suspension under a light microscope after staining with
Figure 6 Effect of active caspase 8, Granzyme B and HIV protease on z-
IETD-AFC. The caspase 8 autoactivation cleavage site fluorogenic substrate
z-IETD-AFC was incubated with recombinant active caspase 8, Granzyme B,
or with either 0.1 or 1 mg of HIV protease as indicated, and fluorescence
measured every 2 min for 30 min Figure 7 Extracts from Jurkat T cells, JB6 or I9.2 cells were treated with HIV-
1 protease, and analysed for caspase 8, caspase 3 and BID cleavage. PCNA
is used as an internal control
Cell Death and Differentiation
HIV-1 protease activates caspase 8
ZNieet al
1179
Trypan blue. The nuclei were removed by the centrifugation at
10006gfor 10 min at 48C. Protein concentrations were determined
with BCA protein assay kit (Pierce Chemical Co, Rockford, IL, USA).
JB6 cells and I9.2 cells which are procaspase 8 deficient T cell
derivatives, were a kind gift of Dr. S Nagata
73
and Dr. J Blenis
74
respectively. JB6 and I9.2 cells were handled in an identical manner to
the method described for Jurkat T cells above.
Preparation of HeLa nuclei
HeLa cell (ATCC, Rockville, MD, USA) nuclei isolation was performed
as described.
75
Nuclei were freshly prepared for each experiment from
the 80% confluent cultures of HeLa cells. Cells were washed three
times with ice-cold PBS (pH 7.4), followed by a single wash with ice-
cold nuclear buffer (10 mM PIPES, 80 mM KCL, 20 mM NaCl, 250 mM
sucrose, 5 mM EGTA, 1 mM DTT, 0.5 mM spermidine, 0.2 mM
spermine, 1 mg ml protease inhibitors, pH 7.4). The cell pellet was
resuspended with two volumes of ice-cold nuclear buffer. The cells
were disrupted with 50 strokes of B-type pestle and 495% lysis
confirmed by Trypan blue exclusion. Nuclei were pelleted (10006gfor
10 min at 48C) and washed twice with nuclear washing buffer (10 mM
PIPES, 10 mM KCl, 2 mM MgCl
2
,1mMDTT,10mM cytochalasin B,
1mgml
71
protease inhibitors, pH 7.4).
HIV-1 protease treatment of cytosolic extracts
Cell extracts treated with HIV-1 protease were carried out in 100 ml
cell-free reaction buffer (complete caspase buffer supplemented with
10 mM phosphocreatine, 2 mM ATP and 150 mg/ml creatine
phosphokinase). The concentration ratio of cytosol proteins and
HIV-1 protease was 1000 : 1. The final concentration of HIV-1 protease
was between 0. 5 ± 1 mg per reaction mixture. HIV-1 protease was
purchased (Bachem Bioscience Inc - King of Prussia, PA, USA) with a
specific activity of 1.81610
4
mmole/min/mg at 378C, with a purity of
496% by SDS ± PAGE and a single peak by RP ± HPLC. Where
indicated, the HIV-1 protease inhibitors (HIV-1-PI) Saquinavir 10 mM
(Roche Laboratories, Mississauga, Ontario, used for data described in
Figures 3 and 4) or Nelfinavir 7 mM (Agouron Laboratories,
Mississauga, Ontario, Canada), used for data described in all Figures
except 3 and 4) were used. Where indicated the human aspartyl
protease renin (Sigma Aldrich Canada Ltd, Oakville, ON, Canada) was
used as a control. Renin substrate 1 (Molecular Probe Inc., Eugene,
OR, USA) was used to measure renin activity in cytosol mixtures
according to the supplied protocol. z-IETD-fmk (Enzyme Systems
Products, Livermore, CA, USA) was used in some experiments as
indicated, at 100 mM dissolved in DMSO (Sigma, Irvine, UK).
Nuclei incubation with HIV-1 protease treated
cytoplasmic extracts
First, a mixture of cytoplasmic extracts and HIV-1 protease were
incubated at 308C for 4 h in cell-free reaction buffer. Then, aliquots of
20 ml HIV-1 protease treated cytoplasmic were incubated with 80 mlof
HeLa cell nuclei (5610
6
nuclei) at 378C in nuclear apoptosis buffer
(nuclear washing buffer supplemented with 2 mM ATP and 5 mM
EGTA). Apoptotic nuclei were determined by Hoechst staining and
DNA fragmentation assay.
Hoechst staining
HeLa nuclei were stained with Hoechst 33342 (Molecular Probes,
Eugene, OR, USA) as previously described
67
in fixing buffer (10%
formaldehyde, 50% glycerol, 100 mM NaCl, 2 mM KCl, 1 mM MgCl2,
0.1 mM EDTA, 1 mg/ml Hoechst 33342, 5 mM HEPES, pH 7.8). The
stained nuclei were imaged under fluorescence microscopy (Zeiss
AxioCAM, Jena, Germany).
DNA fragmentation assay
The DNA fragmentation assay was performed as described.
76
Briefly
2±5610
6
nuclei were pelleted for 20 min at 48C, and dispersed in
Figure 8 Jurkat T cells were infected or mock infected (HIV-) with HIV
IIIb
,in
the presence or absence of z-IETD-fmk, z-VAD-fmk or z-DEVD-fmk, and
assessed for viability (A). Infected cells harvested on day 8 were then
analyzed for caspase 8 and PARP cleavage (B), or for HIV protease
expression (C). HIV protease expression and PARP cleavage were also
assessed in bulk PBL from HIV positive or negative patients, as indicated (D)
Cell Death and Differ entiati on
HIV-1 protease activates caspase 8
ZNieet al
1180
30 ml of lysis buffer (10 mM Tris, 100 mM NaCl, 25 mM EDTA, 0.5%
Sarkosyl) by gentle vortexing. Forty micrograms protease K (Qiagen
Inc., Mississauga, ON, Canada) was added and incubated at 528C
overnight. Then, 40 mg RNase (Sigma, Irvine, UK) was added and
incubated for 2 h at room temperature. The fragmented DNA in the
lysates was detected by 2% agarose gel electrophoresis.
SDS ± PAGE and Western blot
For Western blot analysis, 50 ± 200 mg of cytosolic proteins were
fractionated on 4 ± 15% gradient polyacrylamide gels (Biorad
Laboratories Canada Inc., Hercules, CA, USA), then transferred
onto PVDF membranes (Millipore, Bedford, MA, USA) for 1 h at
100 V using transfer buffer (25 mM Tris, 192 mM glycine, 20%
methanol). The membranes were blocked by incubation in TBS
buffer (20 mM Tris, 500 mM NaCl, 0.05% Tween, pH 7.5)
containing 5% milk for overnight at 48Cor2hatroom
temperature and washed five times with TBS buffer. Then, the
membranes were blotted for 1 h at room temperature with the
various dilutions of primary antibodies, specifically, monoclonal
anti-caspase 8 (Biosource International, Camarillo, CA, USA), anti-
caspase 9 (Medical & Biological Laboratories Co., Watertown, MA,
USA), anti-cytochrome c(BD Pharmingen, Mississauga, ON,
Canada), anti-PARP (Oncogene, Darmstadt, Germany) and anti-
Bcl-2 (Calbiochem, La Jolla, CA, USA), anti-PCNA (Santa Cruz
Biotechnology, Santa Cruz, CA, USA), rabbit anti-caspase 3
77
and
rabbit anti-cFLIP (Alexis Biochemicals, San Diego, CA, USA), goat
anti-BID, anti-actin and anti-DFF45 (Santa Cruz Biotechnology).
The blots were washed five times with TBS and developed with
HRP linked secondary antibodies, sheep anti-mouse Ig, donkey
anti-rabbit Ig (Amersham Pharmacia Biotech, Oakville, ON,
Canada) and anti-goat IgG (Santa Cruz Biotechnology). All the
blots were developed by SuperSignal (Pierce, Rockford, IL, USA),
an enhanced chemiluminescence method, following the manufac-
turer's protocol.
Generation of recombinant caspase 3 and
caspase 8
GST-caspase 8 was made by subcloning full-length cDNA caspase 8
into pGEX-4T-1 (Amersham Pharmacia Biotech) and expression of
GST ±Caspase 8 performed by IPTG stimulation at 308C, according to
the manufacturers instructions, in the presence of 100 uM EGTA and
EDTA. The human caspase 3 cDNA was amplified by RT ± PCR with
the following primers: 5-GATGGAGAACACTGAAAAACTC-3 and 5-
ATCCAACCAACCATTTCTTTAGTG-3 from Jurkat total RNA and
subcloned into BamHI and EcoRI sites of pBSKS+(Stratagene, Cedar
Creek, TX, USA) and sequenced. To produce recombinant caspase-3,
the cysteine 163 of the active site was mutated to serine in order to
avoid autocatalysis. The mutagenesis was performed by overlapping
PCR using PBSKS+caspase-3 as the template, and the mutation was
then confirmed after cloning and sequencing of the PCR product. The
caspase-3-C163S was then subcloned into pGEX2TK (Amersham
Pharmacia Biotech) and transformed into DH5 alpha. Purified
Caspase 3 was made as previously described, followed by removal
of the GST tag by thrombin digestion.
77
Figure 9 Putative role of HIV protease in HIV pathogenesis
Cell Death and Differentiation
HIV-1 protease activates caspase 8
ZNieet al
1181
Cleavage reactions of recombinant caspases
Reactions to assess the ability of HIV-1 protease to cleave
recombinant caspases were performed under the following
conditions: 3 ml of purified recombinant GST-caspase 8 or caspase
3 were mixed with 10 ml of HIV-1 protease buffer (100 mM Na
acetate, 1 mM EDTA, 1 M NaCl, 1 mM DTT, 1 mg/ml BSA pH 4.7)
in the absence or presence of 0.5 mg HIV-1 protease (2 ml)
preincubated for 15 min at room temperature with either 2 mlof
methanol, or 2 ml of 10 mM Saquinavir in methanol. In the case of
caspase 3, Granzyme B (Enzyme Systems, Livermore, CA, USA)
was used as a positive control for cleavage, at the indicated
concentrations. The final reaction mixtures were incubated for the
indicated times at 378C. Cleavage products were then analyzed by
Western blot analysis.
Cytochrome
c
release assay
Cytochrome crelease assay was modified according to a previous
publication.
78
Crude cell extracts were supplemented with an ATP
regenerating system (10 mM phosphocreatine, 2 mM ATP and
150 mg/ml creatine phosphokinase). At various time points, HIV-1
protease treated cytosols were harvested and centrifuged twice at
15 000 g(48C) for 15 min to fractionate the cytosolic (supernatant)
fraction from the mitochondrial pellet. Aliquots of 20 ml cytosolic
protein (200 mg) were separated by 4 ± 15% gradient SDS ± PAGE and
probed with monoclonal antibody against cytochrome c. As indicated,
recombinant active caspase 8 (Biomol, Plymouth Meeting, PA, USA)
or Granzyme B (Enzyme Systems Products, Livermore, CA, USA)
were used as positive controls.
Caspase inhibitors
The caspase consensus site inhibitors z-DEVD-fmk, z-IETD-fmk and
z-VAD-fmk were purchased from Enzyme Systems. Independent
experiments were performed to validate the inhibitory effects of z-
DEVD-fmk, z-IETD-fmk or z-VAD-fmk on caspase activation. Jurkat T
cells were stimulated with recombinant leucine zipper Fas Ligand
(10 g/ml, Immunex) for 6 h at 378C in the absence or presence of z-
DEVD-fmk, z-IETD-fmk or z-VAD-fmk (Enzyme Systems), at con-
centrations ranging from 3 to 300 mM. Each inhibitor blocked
recombinant Fas Ligand (Immunex Corp, Seattle, WA, USA) induced
cell death at all concentrations, in a dose dependant manner (data not
shown).
Cells and HIV infection
Jurkat T cells were purchased from ATCC and maintained in RPMI
medium supplemented with 10% fetal calf serum (FCS, GIBCO).
For experiments using patient peripheral blood lymphocytes (PBL),
consenting patients or controls donated 20 mls of blood into
heparinized tubes, and PBLs extracted using ficol hypaque density
gradient centrifugation, and plastic adherence.
56
Resultant PBL
were cultured in RPMI 1640-10% human AB serum, supplemented
with penicillin/streptamycin and glutamine (Gibco). HIV infection
using HIV IIIb (NIH AIDS Reference Reagent Program) was
performed as previously described;
79
briefly virus containing
supernatants (or mock infected supernatants) were propogated in
PBMC from HIV uninfected donors. Cells are infected by overnight
culture with virus containing (or mock) supernatant (
45
373 pg of
p24/ml). Cell viability following infection was assessed by Trypan
blue exclusion.
Fluorogenic release assays
To assess the activity of different enzymes against z-IETD-AFC
(Enzyme Systems), caspase 8 (Enzyme Systems), 180 mgof
Granzyme B, 0.1 or 1.0_g of HIV protease were added to either
caspase 8 buffer (100 mM HEPES, pH 7.5, 10% v/v sucrose, 10 mM
DTT, 0.5 mM EDTA),
80
Granzyme B buffer (50 mM HEPES, pH 7.4,
0.1% CHAPS, 0.1 M NaCl, 10% v/v sucrose, 10 mM DTT)
81
or to HIV
protease buffer (100 mM Na Acetate, 4 mM EDTA, 300 mM NaCl,
pH 4.7)
82
to achieve a final volume of 500 ml. After 30 min, with the
reaction mixture in a fluorimeter (CytoFluor 2300, Millipore) adjusted to
400 nm excitation, 505 nm emission, 20 mlofz-IETD-AFC(20mM
stock) was added, and fluorescence release measured every 2 min
until 30 min.
82
Data presented representative of results obtained using
all three buffers. Independent experiments using the HIV protease
fluorogenic substrate. DABCYL-_-Abu-Ser-Gln-Asn-Tyr-Pro-Ile-Val-
Gln-EDAN (Bachem, King of Prussia PA, USA) were performed with
each buffer to confirm the activity of HIV protease under these
conditions (data not shown).
Acknowledgements
Informed consent was obtained from all subjects prior to blood collection,
and the study was reviewed and approved by the ethics committee of the
Ottawa Hospital/University of Ottawa. The authors gratefully acknowl-
edge the helpful discussions and advice of Dr. BWD Badley, as well as
the administrative expertise of A Carisse. BN Phenix and JJ Lum are
supported by a Studentship award from the Ontario HIV Treatment
Network (OHTN). AD Badley is supported by an OHTN Career Scientist
Award. This work is supported by grants from the Doris Duke Foundation
(#T98026), the Canadian Institute of Health Research (#HOP-36047),
the Canadian Foundation for AIDS Research and a Premier's Research
Excellence Award.
References
1. Badley AD, McElhinny JA, Leibson PJ, Lynch DH, Alderson MR and Paya CV
(1996) Upregulation of Fas ligand expression by human immunodeficiency virus
in human macrophages mediates apoptosis of uninfected T lymphocytes. J.
Virol. 70: 199 ± 206
2. Badley AD, Dockrell D, Simpson M, Schut R, Lynch DH, Leibson P and Paya CV
(1997) Macrophage-dependent apoptosis of CD4+T lymphocytes from HIV-
infected individuals is mediated by FasL and tumor necrosis factor. J. Exp. Med.
185: 55 ± 64
3. Banda NK, Bernier J, Kurahara DK, Kurrle R, Haigwood N, Sekaly RP and Finkel
TH (1992) Crosslinking CD4 by Human Immunodeficiency virus gp120 primes T
cells for activation-induced apoptosis. J. Exp. Med. 176: 1099 ± 1106
4. Cottrez F, Manca F, Dalgleish AG, Arenzana-Seisdedos F, Capron A and Groux
H (1997) Priming of human CD4+antigen-specific T cells to undergo apoptosis by
HIV-infected monocytes. J. Clin. Invest. 99: 257± 266
5. Herbein G, Van Lint C, Lovett JL and VerdinE (1998) Distinct mechanisms trigger
apoptosis in human immunodeficiency virus type-1 infected and in uninfected
bystander T lymphocytes. J. Virol. 72: 660 ± 670
6. Kameoka M, Suzuki S, Kimura T, Fujinaga K, Auwanit W, Luftig RB and Ikuta K
(1997) Exposure of resting peripheral blood T cells to HIV-1 particles generates
CD25+killer cells in a small subset, leading to induction of apoptosis in bystander
cells. Int. Immunol. 9: 1453 ± 1462
7. Nardelli B, Gonzalez CJ, Schechter M and Valentine FT (1995) CD4+blood
lymphocytes are rapidly killed in vitro by contact with autologous human
immunodeficiency virus-infected cells. Proc. Natl. Acad. Sci. USA. 92: 7312 ±
7316
Cell Death and Differ entiati on
HIV-1 protease activates caspase 8
ZNieet al
1182
8. Orlikowsky T, Wang Z-Q, Dudhane A, Horowitz H, Riethmuller G and Hoffman
MK (1997) Cytotoxic monocytes in the blood of HIV type-1 infected subjects
destroy targeted T cells in a CD-95-dependent fashion. AIDS Res. Hum. Retro.
13: 953 ± 960
9. Mitra D, Steiner M, Lynch DH, Staiano-Coico L and Laurence J (1996) HIV-1
upregulates Fas ligand expression in CD4+Tcells in vitro and in vivo: association
with Fas-mediated apoptosis and modulation by aurintricarboxylic acid.
Immunol. 87: 581 ± 585
10. Sloand EM, Young NS, Kumar P, Weichold FF, Sato T and Maciejewski JP
(1997) Role of Fas ligand and receptor in the mechanism of T-cell depletion in
acquired immunodeficiency syndrome: effect on CD4+lymphocyte depletion
and human immunodeficiency virus replication. Blood 89: 1357 ±1363
11. Alderson MR, Tough TW, Davis-Smith T, Braddy S, Falk B, Schooley A, Goodwin
RG, Smith CA, Ramsdell F and Lynch DH (1995) Fas ligand mediates activation-
induced cell death in human T lymphocytes. J. Exp. Med. 181: 71 ± 77
12. Dhein J, Walczak H, Baumler C, Debatin KM and Krammer PH (1995) Autocrine
T-cell suicide mediated by APO-1/(Fas/CD95). Nature 373: 438 ± 443
13. Ju ST, Panka DJ, Cui H, Ettinger R, El-Khatib M, Sherr DH, Stanger BZ and
Marshak-Rothstein A (1995) Fas (CD95) FasL interactions required for
programmed cell death after T-cell activation. Nature 373: 444 ±448
14. Wesselborg S, Janssen O and Kabelitz D (1993) Induction of activation-driven
death (apoptosis) in activated but notresting peripheral blood T cells. J. Immunol.
150: 4338 ± 4345
15. Gandhi RT, Chen BK, Straus SE, Dale JK, Lenardo MJ and Baltimore D (1998)
HIV-1 directly kills CD4+T cells by a Fas-independent mechanism. J. Exp. Med.
187: 1113 ± 1122
16. Katsikis PD, Garcia-Ojeda ME, Wunderlich ES, Smith CA, Yagita H, Okumura K,
Kayagaki N, Alderson M, Herzenberg LA and Herzenberg LA (1996) Activation-
induced peripheral blood T cell apoptosis is Fas independent in HIV-infected
individuals. Int. Immunol. 8: 1311 ± 1317
17. Katsikis PD, Garcia-Ojeda ME, Torres-RocaJF, Tijoe IM, Smith CA, Herzenberg
LA and Herzenberg LA (1997) Interleukin-1B converting enzyme-like protease
involvement in Fas-induced and activation-induced peripheral blood T cell
apoptosis in HIV infection. TNF-related apoptosis-inducing ligand can mediate
activation-induced T cell death in HIV infection. J. Exp. Med. 186: 1365± 1372
18. Ledru E, Lecoeur H, Garcia S, Debord T and Gougeon ML (1998) Differential
susceptibility to activation-induced apoptosis among peripheral Th1 subsets:
correlation with Bcl-2 expression and consequences for AIDS pathogenesis. J.
Immunol. 160: 3194 ± 3206
19. Groux H, Torpier G, Monte D, Mouton Y, Capron A and Ameisen JC (1992)
Activation-induced death by apoptosis in CD4+T cells from human
immunodeficiency virus-infected asymptomatic individuals. J. Exp. Med. 175:
331 ± 340
20. Algeciras A, Dockrell DH, Lynch DH and Paya CV (1998) CD4 regulates
susceptibility to Fas ligand - and tumor necrosis factor-mediated apoptosis. J.
Exp. Med. 187: 711 ± 720
21. Laurent-Crawford AG, Krust B, Riviere Y, Desgranges C, Muller S, Kieney MP,
Dauguet C and Hovanessian AG (1993) Membrane expression of HIV envelope
glycoproteins triggers apoptosis in CD4 cells. AIDS Res. Hum. Retro. 9: 761 ±
773
22. Oyaizu N, McCloskey TW, Coronesi M, Chirmule N, Kalyanaraman VS and
Pahwa S (1993) Accelerated apoptosis in peripheral blood mononuclear cells
(PBMCs) from human immunodeficiency virus type-1 infected patients and in
CD4 cross-linked PBMCs from normal individuals. Blood 82: 3392 ± 3400
23. Jeremias I, Herr I, Boehler T and Debatin KM (1998) TRAIL/Apo-2-ligand-
induced apoptosis in human T cells. Eur. J. Immunol. 28: 143± 152
24. Glynn JM, McElligott DL and Mosier D (1996) Apoptosis induced by HIV infection
in H9 T cells is blocked by ICE-Family protease inhibition but not by a Fas (CD95)
antagonist. J. Immunol. 157: 2754 ± 2758
25. Noraz N, Gozlan J, Corbeil J, Brunner T and Spector SA (1997) HIV-induced
apoptosis of activated primary CD4+T lymphocytes is not mediated by Fas ± Fas
ligand. AIDS 11: 1671 ±1680
26. Li CJ, Friedman DJ, Wang C, Metelev V and Pardee AB (1995) Induction of
apoptosis in uninfected lymphocytes by HIV-1 Tat protein. Science 268: 429 ±
431
27. New DR, Maggirwar SB, Epstein LG, Dewhurst S and Gelbard HA (1998) HIV-1
Tat induces neuronal death via tumor necrosis factor-aand activation of non-N-
methyl-D-aspartate receptors by a NF
k
B-independent mechanism. J. Biol.
Chem. 273: 17852 ± 17858
28. Patki AH and Lederman MM (1996) HIV-1 Tat protein and its inhibitor Ro 24-7429
inhibit lymphocyte proliferation and induce apoptosis in peripheral blood
mononuclear cells from healthy donors. Cell Immunol. 169: 40 ± 46
29. Fujii Y, Otake K, Tashiro M and Adachi Y (1996) Human immunodeficiency virus
type 1 Nef protein on the cell surface is cytocidal for human CD4+T cells. FEBS
Letters 393: 105 ± 108
30. Fujii Y, Otake K, Tashiro M and Adachi Y (1996) Soluble Nef antigen of HIV-1 is
cytotoxic for human CD3+T cells. FEBS Letters 393: 93± 96
31. FujiiY, Otake K, Tashiro M andAdachi Y (1996) In vitro cytocidal effects of human
immunodeficiency virus type 1 Nef on unprimed human CD4+T cells without
MHC restriction. J. Gen. Virol. 77: 2943 ± 2951
32. Hanna Z, Kay DG, Rebai N, Guimond A, Jothy S and Jolicoeur P (1998) Nef
harbors a major determinantof pathogenicityfor an AIDS-like disease induced by
HIV-1 in transgenic mice. Cell 95: 163 ± 175
33. Poon B, Grovit-Ferbas K, Stewart SA and Chen ISY (1998) Cell cycle arrest by
vpr in HIV-1 virions and insensitivity to antiretroviral agents. Science 281: 266 ±
269
34. Stewart SA, Poon B, Jowett JBM and Chen ISY (1997) Human immunodefi-
ciency virus type 1 vpr induces apoptosis following cell cycle arrest. J. Virol. 71:
5579 ± 5592
35. Yao X-J, Mouland AJ, Subbramanian RA, Forget J, Rougeau N, Bergeron D and
Cohen EA (1998) Vpr stimulates viral expression and induces cell killing in
human immunodeficiency virus type 1-infected dividing jurkat T cells. J. Virol. 72:
4686 ± 4693
36. Adams LD, Tomasselli AG, Robbins P, Moss P and Heinrikson RL (1992) HIV-1
protease cleaves actin during acute infection human T-lymphocytes. AIDS Res.
Hum. Retro. 8: 291 ± 295
37. Buttner J, Dornmair K and Schramm HJ (1997) Screening of inhibitors of HIV-1
protease using an Escherichia coli cell assay. Biochem. Biophy. Res. Commun.
233: 36±38
38. Konvalinka J, Litterst MA, Welker R, Kottler H, Rippman F, Heuser A-M and
Krausslich H-G (1995) An active site mutation in the HIV type 1 proteinase (PR)
causes reduced PR activity and loss of PR mediated cytotoxicity without
apparent effect on virus maturation and infectivity. J. Virol. 69: 7180± 7186
39. Rivi re Y, Blank V, Kourilsky P and Israel A (1991) Processing of the precursor of
NF-kappa B by the HIV-1 protease during acute infection. Nature 350: 625 ± 626
40. Shimizu T and Pommier Y (1996) DNA fragmentation induced by protease
activation in p53-null human leukemia HL60 cells undergoing apoptosis
following treatment with the topoisomerase 1 inhibitor campothecin: cell-free
system studies. Exper. Cell. Res. 226: 292 ± 301
41. Kaplan AH and Swanstrom R (1991) The HIV-1 gag precursor is processed via
two pathways: implications for cytotoxicity. Biomed. Biochim. Acta. 50: 647 ± 653
42. Kaplan AH and Swanstrom R (1991) Human immunodeficiency virus type 1 Gag
proteins are processed in two cellular compartments. Proc. Natl. Acad. Sci. USA.
88: 4528 ± 4532
43. Strack PR, West Frey M, Rizzo CJ, Cordova B, George HJ, Meade R, Ho W,
Corman J, Tritch R and Korant BD (1996) Apoptosis mediated by HIV proteaseis
preceded by cleavage of Bcl-2. Proc. Natl. Acad. Sci. USA. 93: 9571 ± 9576
44. Liu X, Kim CN, Yang J, Jemmerson R and Wang R (1996) Induction of apoptotic
program in cell-free extracts: requirement for dATP and cytochrome c.Cell86:
147 ± 157
45. Cosulich SC, Green S and Clarke PR (1996) Bcl-2 regulates activation of
apoptotic proteases in a cell-free system. Curr. Biology 6: 997± 1005
46. Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, Newmeyer DD, Wang H,
Reed JC, Nicholson DW, Alnemri ES, Green DR and Martin SJ (1999) Ordering
the cytochrome c-initiated caspase cascade: hierarchical activation of caspase-
2,-3,-6,-7,-8, and -10 in a caspase 9-dependent manner. J. Cell. Biol. 144: 281 ±
292
47. Muzio M, Salveson GS and Dixit VM (1997) FLICE induced apoptosis in a cell-
free system. J. Biol. Chem. 272: 2952 ± 2956
48. Kroemer G and Reed JC (1998) Mitochondrial control of cell death. Nat. Med. 6:
513 ± 519
49. Green DR and Reed JC (1998) Mitochondria and apotposis. Science 281:
1309 ± 1312
50. Saleh A, Srinivasula SM, Acharya S, Fishel R and Alnemri ES (1999)
Cytochrome c and dATP-mediated oligomerization of Apaf-1 is a prerequisite
for procaspase-9 activation. J. Biol. Chem. 274: 17941± 17945
Cell Death and Differentiation
HIV-1 protease activates caspase 8
ZNieet al
1183
51. Hu Y, Benedict MA, Ding L and Nunez G (1999) Role of cytochrome c and dATP/
ATP hydrolysis in Apaf-1-mediated caspase-9 activation and apoptosis. EMBO
J. 18: 3586 ± 3595
52. Li H, Zhu H, Xu CJ and Yuan J (1998) Cleavage of BID by caspase 8 meidates the
mitochondrial damage in the Fas pathway of apoptosis. Cell 94: 491 ± 501
53. Wei MC, Lindstein T, Mootha VK, Weiler S, Gross A, Ashiya M, Thompson CB
and Korsmeyer SJ (2000) tBID, a membrane-targeted death ligand, oligomerizes
BAK to release cytochrome c. Genes Dev. 14: 2060 ± 2071
54. Tomasselli AG, Hui JO, LA, Chosay J, Lowery D, Greenberg B, Yem A, Deibel
MR, Zurcher-Neely H and Heinrickson RL (1991) Actin, troponin C, alzheimer
amyloid precursor protein and pro-interleukin 1b as substrates of the protease
from human immunodeficiency virus. J. Biol. Chem. 266: 14548 ±14553
55. Krammer PH (1999) CD95 (APO-1/Fas)-meidated apoptosis: live and let die.
Adv. Immunol. 71: 163 ±210
56. Medema JP, Scaffidi C, Kischek FC, Shevchenko A, Mann M, Krammer PH and
Peter ME (1997) FLICE is activated by association with the CD95 death-inducing
signaling complex (DISC). EMBO J. 16: 2794 ± 2804
57. Andrade F, Roy S, Nicholson D, Thornberry N, Rosen A and Casciola-Rosen L
(1998) Granzyme B directly and efficiently cleaves several downstream caspase
substrates: implications for CTL-induced apoptosis. Immunity 8: 451 ±460
58. Chou KC and Zhang CT (1993) Studies on the specificity of HIV protease: an
application of Markov chain theory. J. Protein Chem. 12: 709± 724
59. Tewari M, Quan LT, O'Rourke K, Desnoyers S, Zeng Z, Beidler DR, Poirier GG,
Salvesen GS and Dixit VM (1995) Yama/CPP32 beta, a mammalian homolog of
CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly
(ADP-ribose) polymerase. Cell. 81: 801 ± 809
60. Nicholson DW, Ali A, Thornberry N, Vaillancourt JP, Ding CK, Gallant M, Gareau
Y, Griffin PR, Labelle Mand Lazebnik YA (1995) Identification and inhibition of the
ICE/CED-2 protease necessary for mammalian apoptosis. Nature 376: 37 ±43
61. Badley AD, Pilon AA, Landay A and Lynch DH (2000) Mechanisms of HIV
associated lymphocyte apoptosis. Blood 96: 2951 ± 2964
62. Barry MG and McFadden G (1998) Apoptosis regulators from DNA viruses. Curr.
Opin. Immunol. 10: 422 ±430
63. Thome M, Schnelder P, Hofmann K, Foickenscher H, Neinl E, Neipel F,
MattmannC,BurnsK,BodmerJL,SchroterM,ScaffidiC,KrammerPH,PeterME
and Tschopp J (1997) Viral FLICE-inhibitory proteins (FLIPs) prevent apoptosis
induced by death receptors. Nature 386: 517 ±521
64. HonerB, Shoeman RL and Traub P (1991) Human immunodeficiency virus type 1
protease microinjected into cultured humanskin fibroblasts cleaves vimentin and
affects cytoskeletal and nuclear architecture. J. Cell. Sci. 100: 799± 807
65. Shoeman RL, Honer B, Stoller TJ, Kesselmeier C, Miedel MC, Traub P and
Graves MC (1990) Human immunodeficiency virus type 1 protease cleaves the
inermediate filament proteins vimentin, desmin, and glial fibrillary acidic protein.
Natl. Acad. Sci. USA. 87: 6336 ± 6340
66. Korant B, Strack P, Frey MW and Rizzo CJ (1998) A cellular anti-apoptosis
protein is cleaved by the HIV-1 protease. Adv. Exp. Med. Biol. 436: 27± 29
67. Martin SJ, Newmeyer DD, Mathias S, Farschon D, Wang H, Reed JC, Kolesnick
RN and Green DR (1995) Cell-free reconstitution of Fas-, UV radiation and
ceramide-induced apoptosis. EMBO J. 14: 5191 ± 5200
68. Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, Bodmer JL,
Schroter M, Burns K, Mattmann C, Rimoldi D, French LE and Tschopp J (1997)
Inhibition of death receptor signals by cellular FLIP. Nature 388: 190 ± 195
69. Scaffidi C, Schmitz I, Zha J, Korsmeyer SJ, Krammer PH and Peter ME (1999)
Differential modulation of apoptosis sensitivity in CD95 type I and type II cells. J.
Biol. Chem. 274: 22532 ± 22538
70. Flexner C (1998) HIV-protease inhibitors. N. Engl. J. Med. 338: 1281± 1292
71. Phenix BN, Lum JJ, Sanchez-Dardon J and Badley AD (2001) Antiapoptotic
mechanism of HIV protease inhibitors: preventing mitochondrial potential loss.
Blood 98: 1078 ± 1085
72. Stennicke HR and Salvesen GS (1997) Biochemical characteristics of caspase-
3,-6,-7, and -8. J. Biol. Chem. 272: 25719 ± 25723
73. Kawahara A, Ohsawa Y, Matsumura H, Uchiyama Y and Nagata S (1998)
Caspase-independent cell killing by Fas-associated protein with death domain.
J. Cell. Biol. 143: 1353 ±1360
74. Juo P, Kuo CJ, Yuan J and Blenis J (1998) Essential requirement for caspase-8/
FLICE in the initiation of the Fas- induced apoptotic cascade. Curr. Biol. 8: 1001 ±
1008
75. Wood ER and Earnshaw WC (1990) Mitototic chromatin condensation in vitro
using somatic cell extracts as nuclei with variable levels of endogenous
topoisomerese II. Cell. Biol. 111: 2389 ± 2850
76. Zhu N and Wang Z (1997) An assay for DNA fragmentation in apoptosis without
phenol/chloroform extraction and ethanol precipitation. Analytical Biochemistry
246: 155 ± 158
77. Alam A, Braun MY, Hartgers F, Lesage G, Cohen L, HugoP, Denis F and Sekaly
R-P (1997) Specific activation of the cysteine protease CPP32 during the
negative selection of T cells in the thymus. J. Exp. Med. 186: 1503 ± 1512
78. Evans EK, Kuwana T, Strum SL, Smith JJ, Newmeyer DD and Kornbluth S (1997)
Reaper-induced apoptosis in a vertebrate system. EMBO J. 16: 7372± 7381
79. Schmitz I, Walczak H, Krammer PH and Peter ME (2000) The two CD95
apoptosis signaling pathways may be a way of cells to respond to different
amounts and/or forms of CD95 ligand produced in different tissues. Cell Death
and Diff. 7: 756 ± 758
80. Stennicke HR and Salvesen GS (1999) Catalytic properties of the caspases. Cell
Death Differ. 6: 1054 ± 1059
81. Muzio M, Chinnaiyan AM, Kischkel FC,O'Rourke K, Shevchenko A, Ni J, Scaffidi
C, Bretz JD, Zhand M, Gentz R, Mann M, Krammer PH, Peter ME and Dixit VM
(1996) FLICE, a novel FADD/homologous ICE/CED-3-like protease, is recruited
to the CD95 (Fas/APO-1) death-inducing signaling complex. Cell. 85: 817± 827
82. Griffiths JT, Phylip LH, Konvalinka J, Strop P, Gustchina A, Wlodawer A,
Davenport RJ, Briggs R, Dunn BM and Kay J (1992) Different requirements for
productive interaction between the active site of HIV-1proteinase and substrates
containing - hydrophobic*hydrophobic- or -aromatic*pro- cleavage sites.
Biochemistry 31: 5193 ± 5200
Cell Death and Differ entiati on
HIV-1 protease activates caspase 8
ZNieet al
1184
... Given the emergence of drug-resistant strains of HIV-1, novel strategies of targeting HIV PR are urgently needed. Interestingly, HIV PR has been shown to interact with numerous host cell proteins [15][16][17][18]. As the cleavage of certain host substrates by HIV-1 PR has been linked to cell death, targeting the interactions between PR and the host rather than PR and viral polyprotein precursors may open promising new avenues for antiviral therapy. ...
... In a cell-free system using cytoplasmic extracts from Jurkat T cells, HIV-1 PR was shown to cleave and activate pro-caspase-8, an initiator caspase of extrinsic apoptosis [18]. The cleavage of pro-caspase-8 in cell extracts was associated with the cleavage of BID, a member of the BCL-2 family that is known to be cleaved by caspase-8 [42]. ...
... Pro-caspase-8 cleavage was also associated with mitochondrial cytochrome c release, consistent with previous data showing that truncated BID (tBID) translocates into the mitochondria and induces cytochrome c release into the cytosol [43]. Notably, the HIV-1 PR cleavage site of pro-caspase-8 differs from the typical cleavage site, as a mutation of the normal procaspase-8 cleavage site did not affect cleavage patterns by HIV-1 PR [18]. A follow-up study showed that transfection of the novel fragment produced by HIV-1 PR cleavage of pro-caspase-8 (termed casp8p41) into both HeLa cells and primary CD4 + T cells caused apoptosis [44]. ...
Article
Full-text available
HIV-1 protease (PR) is a viral enzyme that cleaves the Gag and Gag-Pol polyprotein precursors to convert them into their functional forms, a process which is essential to generate infectious viral particles. Due to its broad substrate specificity, HIV-1 PR can also cleave certain host cell proteins. Several studies have identified host cell substrates of HIV-1 PR and described the potential impact of their cleavage on HIV-1-infected cells. Of particular interest is the interaction between PR and the caspase recruitment domain-containing protein 8 (CARD8) inflammasome. A recent study demonstrated that CARD8 can sense HIV-1 PR activity and induce cell death. While PR typically has low levels of intracellular activity prior to viral budding, premature PR activation can be achieved using certain non-nucleoside reverse transcriptase inhibitors (NNRTIs), resulting in CARD8 cleavage and downstream pyroptosis. Used together with latency reversal agents, the induction of premature PR activation to trigger CARD8-mediated cell killing may help eliminate latent reservoirs in people living with HIV. This represents a novel strategy of utilizing PR as an antiviral target through premature activation rather than inhibition. In this review, we discuss the viral and host substrates of HIV-1 protease and highlight potential applications and advantages of targeting CARD8 sensing of HIV-1 PR.
... (A) The viral integration itself can activate DNA-dependent protein kinase (DNA-PK), resulting in phosphorylation of both p53 and H2AX and triggering cell death(13). (B) CD4 1 T cells abortively infected with HIV-1 can die through caspase-1-dependent pyroptosis(14)(15)(16). (C) HIV protease cleaves the cellular protein procaspase-8 to generate a novel caspase-8 fragment, termed Casp8p41(97,98), which, after translocation to the mitochondria, induces depolarization of the mitochondrial outer membrane (MOMP), resulting in release of cytochrome c and activation of caspase-3 (99) in a Bax/Bak-and procaspase-9-dependent manner (100). (D) HIV-infected cells can undergo necrosis with an autophagic component ...
Article
Full-text available
HIV-1 (HIV) infects CD4+ T cells, the gradual depletion of which can lead to AIDS in the absence of antiretroviral therapy (ART). Some cells, however, survive HIV infection and persist as part of the latently infected reservoir that causes recurrent viremia after ART cessation. Improved understanding of the mechanisms of HIV-mediated cell death could lead to a way to clear the latent reservoir. Death induced by survival gene elimination (DISE), an RNA interference (RNAi)-based mechanism, kills cells through short RNAs (sRNAs) with toxic 6-mer seeds (positions 2 to 7 of sRNA). These toxic seeds target the 3' untranslated region (UTR) of mRNAs, decreasing the expression of hundreds of genes critical for cell survival. In most cells under normal conditions, highly expressed cell-encoded nontoxic microRNAs (miRNAs) block access of toxic sRNAs to the RNA-induced silencing complex (RISC) that mediates RNAi, promoting cell survival. HIV has been shown to inhibit the biogenesis of host miRNAs in multiple ways. We now report that HIV infection of cells deficient in miRNA expression or function results in enhanced RISC loading of an HIV-encoded miRNA HIV-miR-TAR-3p, which can kill cells by DISE through a noncanonical (positions 3 to 8) 6-mer seed. In addition, cellular RISC-bound sRNAs shift to lower seed viability. This also occurs after latent HIV provirus reactivation in J-Lat cells, suggesting independence of permissiveness of cells to viral infection. More precise targeting of the balance between protective and cytotoxic sRNAs could provide new avenues to explore novel cell death mechanisms that could be used to kill latent HIV. IMPORTANCE Several mechanisms by which initial HIV infection is cytotoxic to infected cells have been reported and involve various forms of cell death. Characterizing the mechanisms underlying the long-term survival of certain T cells that become persistent provirus reservoirs is critical to developing a cure. We recently discovered death induced by survival gene elimination (DISE), an RNAi-based mechanism of cell death whereby toxic short RNAs (sRNAs) containing 6-mer seed sequences (exerting 6-mer seed toxicity) targeting essential survival genes are loaded into RNA-induced silencing complex (RISC) complexes, resulting in inescapable cell death. We now report that HIV infection in cells with low miRNA expression causes a shift of mostly cellular RISC-bound sRNAs to more toxic seeds. This could prime cells to DISE and is further enhanced by the viral microRNA (miRNA) HIV-miR-TAR-3p, which carries a toxic noncanonical 6-mer seed. Our data provide multiple new avenues to explore novel cell death mechanisms that could be used to kill latent HIV.
... Enhancement and killing by VbP were also shown to be dependent on proteasomal degradation (Fig. 3d), a key process to generate the bioactive CARD8C. Caspase 8 has also been shown to be cleaved by HIV-1 protease 33 . Knocking out CASP8 did not diminish killing or enhancement after NNRTI and VbP treatment (Extended Data Fig. 7), suggesting that CASP8 was not involved in NNRTI-mediated cell killing. ...
Article
Full-text available
Non-nucleoside reverse transcriptase inhibitors (NNRTIs) induce pyroptosis of HIV-1-infected CD4⁺ T cells through induction of intracellular HIV-1 protease activity, which activates the CARD8 inflammasome. Because high concentrations of NNRTIs are required for efficient elimination of HIV-1-infected cells, it is important to elucidate ways to sensitize the CARD8 inflammasome to NNRTI-induced activation. We show that this sensitization can be achieved through chemical inhibition of the CARD8 negative regulator DPP9. The DPP9 inhibitor Val-boroPro (VbP) can kill HIV-1-infected cells without the presence of NNRTIs and act synergistically with NNRTIs to promote clearance of HIV-1-infected cells in vitro and in humanized mice. More importantly, VbP is able to enhance clearance of residual HIV-1 in CD4⁺ T cells isolated from people living with HIV (PLWH). We also show that VbP can partially overcome NNRTI resistance. This offers a promising strategy for enhancing NNRTI efficacy in the elimination of HIV-1 reservoirs in PLWH.
... SARS-CoV ORF7a and 7b induce apoptosis by activating caspase 3 (SS et al., 2007). HIV-1 Vpr destroys mitochondrial transmembrane potential and activates both caspases-8 and 9 to induce apoptosis (Jacotot et al., 2000;Nie et al., 2002). HCV NS4A changes the distribution of mitochondria in cells and causes mitochondrial damage, which finally induces apoptosis by activating caspase-3 (Nomura- Takigawa et al., 2006). ...
Article
Full-text available
Porcine sapelovirus (PSV) is the causative pathogen of reproductive obstacles, acute diarrhea, respiratory distress, or severe polioencephalomyelitis in swine. Nevertheless, the pathogenicity and pathogenic mechanism of PSV infection are not fully understood, which hinders disease prevention and control. In this study, we found that PSV was sensitive to type I interferon (IFN-β). However, PSV could not activate the IFN-β promoter and induce IFN-β mRNA expression, indicating that PSV has evolved an effective mechanism to block IFN-β production. Further study showed that PSV inhibited the production of IFN-β by cleaving mitochondrial antiviral signaling (MAVS) and degrading melanoma differentiation-associated gene 5 (MDA5) and TANK-binding kinase 1 (TBK1) through viral 3Cpro. In addition, our study demonstrated that PSV 3Cpro degrades MDA5 and TBK1 through its protease activity and cleaves MAVS through the caspase pathway. Collectively, our results revealed that PSV inhibits the production of type I interferon to escape host antiviral immunity through cleaving and degrading the adaptor molecules.
... Following LPS treatment, caspase-3 activity was determined as previously described with modification [38]. Briefly, 5 × 10 6 cells were lysed using the repeated freezethaw cycles method in 50 μl cell lysis buffer (50 mM HEPES, pH 7.4, 100 mM NaCl, 0.1% CHAPS, 1 mM DTT, 0.1 mM EDTA) in a 96 well plate. ...
Article
Full-text available
Background Acute kidney injury (AKI) is associated with a severe decline in kidney function caused by abnormalities within the podocytes' glomerular matrix. Recently, AKI has been linked to alterations in glycolysis and the activity of glycolytic enzymes, including pyruvate kinase M2 (PKM2). However, the contribution of this enzyme to AKI remains largely unexplored. Methods Cre-loxP technology was used to examine the effects of PKM2 specific deletion in podocytes on the activation status of key signaling pathways involved in the pathophysiology of AKI by lipopolysaccharides (LPS). In addition, we used lentiviral shRNA to generate murine podocytes deficient in PKM2 and investigated the molecular mechanisms mediating PKM2 actions in vitro. Results Specific PKM2 deletion in podocytes ameliorated LPS-induced protein excretion and alleviated LPS-induced alterations in blood urea nitrogen and serum albumin levels. In addition, PKM2 deletion in podocytes alleviated LPS-induced structural and morphological alterations to the tubules and to the brush borders. At the molecular level, PKM2 deficiency in podocytes suppressed LPS-induced inflammation and apoptosis. In vitro, PKM2 knockdown in murine podocytes diminished LPS-induced apoptosis. These effects were concomitant with a reduction in LPS-induced activation of β-catenin and the loss of Wilms’ Tumor 1 (WT1) and nephrin. Notably, the overexpression of a constitutively active mutant of β-catenin abolished the protective effect of PKM2 knockdown. Conversely, PKM2 knockdown cells reconstituted with the phosphotyrosine binding–deficient PKM2 mutant (K433E) recapitulated the effect of PKM2 depletion on LPS-induced apoptosis, β-catenin activation, and reduction in WT1 expression. Conclusions Taken together, our data demonstrates that PKM2 plays a key role in podocyte injury and suggests that targetting PKM2 in podocytes could serve as a promising therapeutic strategy for AKI. Trial registration Not applicable.
... HIV infection of an individual T cell most often results in cell death through a number of proposed pathways, including pyroptosis, innate sensing of HIV proteins or nucleic acids, or apoptosis [16]. One pathway of HIV induced cell death occurs at a post integration step, and involves HIV protease which is active within the infected cell cytosol, cleaving both the HIV polyprotein as well as host proteins including Procaspase 8 [17]. The HIV specific caspase 8 fragment, called Casp8p41, adopts an alpha helical conformation which resembles a BH3 like conformation normally associated with the Bcl2 family of apoptosis regulatory proteins [18]. ...
Article
Full-text available
Background Achieving a functional or sterilizing cure for HIV will require identification of therapeutic interventions that reduce HIV reservoir size in infected individuals. Proteasome inhibitors, such as ixazomib, impact multiple aspects of HIV biology including latency, transcription initiation, viral replication, and infected cell killing through the HIV protease – Casp8p41 pathway, resulting in latency reversal and reduced measures of HIV reservoir size ex vivo. Methods We conducted a phase 1b/2a dose escalating, open label trial of weekly oral ixazomib for 24 weeks in antiretroviral (ART)-suppressed, HIV positive adults (NCT02946047). The study was conducted from March 2017 to August 2019 at two tertiary referral centers in the United States. The primary outcomes were safety and tolerability of oral ixazomib. Secondary outcomes included changes in immunologic markers and estimates of HIV reservoir size after ixazomib treatment. Findings Sixteen participants completed the study. Ixazomib up to 4mg weekly was safe and well-tolerated, yielding no treatment-emergent events above grade 1. In exploratory analyses, ixazomib treatment was associated with detectable viremia that was below the lower limit of quantification (LLQ) in 9 participants, and viremia that was above LLQ in 4 of 16 participants. While treatment was associated with reduced CD4 counts [baseline 783 cells/ mm³ vs. week-24 724 cells/ mm³ p=0.003], there were no changes in markers of cellular activation, exhaustion or inflammation. Total HIV DNA and proviral sequencing were not altered by ixazomib treatment. Intact proviral DNA assay (IPDA) identified intact proviruses in 14 patients pre-treatment, and in 10/14 of those subjects post treatment values were reduced (P=0.068), allowing a calculated intact proviral half life of 0.6 years (95% CI 0.3, 2.5), compared to 7.1 years (95% CI 3.9, 18, p=0.004) in historical controls. Differentiation Quantitative Viral Outgrowth Assays (dQVOA) identified measurable proviruses in 15 subjects pre-treatment; post-treatment values were numerically reduced in 9, but overall differences were not significantly different. Interpretation Our study successfully met its primary endpoint of demonstrating the safety of ixazomib for 24 weeks in HIV infected persons. Exploratory analyses suggest that the effects observed ex vivo of latency reversal and reductions in HIV reservoir size, also occur in vivo. Future controlled studies of ixazomib are warranted. Funding This study was funded by Millennium Pharmaceuticals Inc..; the Mayo Clinic Foundation; the National Institutes of Health, including the National Institute of Allergy and Infectious Diseases, Division of AIDS, the National Heart, Lung and Blood Institute, the National Institute of Diabetes and Digestive and Kidney Diseases, the National Institute of Neurological Disorders and Stroke, and the National Institute on Drug Abuse. Mayo Clinic also acknowledges generous funding support from Mr. Joseph T. and Mrs. Michele P. Betten.
... The serine protease Granzyme B (GzmB) is a potent inducer of apoptosis in target cells when released by cytotoxic T lymphocytes (CTL) or natural killer (NK) cells, representing one of the two dominant mechanisms by which T cells mediate cancer cell death [1,2]. Considering the GzmB's preference for cleaving after aspartic acid, several substrates containing IEPD and IETD sequences coupled to chromogenic or fluorescent leaving groups have been successfully used for GzmB detection [3][4][5]. However, these two sequences lack specificity as they can also be cleaved by some caspases, a family of cysteine proteases that are also activated during apoptosis [6,7]. ...
Conference Paper
Full-text available
The synthesis and characterization of a new fluorogenic substrate for granzyme B (GzmB) is reported. The substrate design was based on the fluorescence resonance energy transfer (FRET) principle using 5-(2′-aminoethyl)aminonaphthalene sulfonic acid (Edans) and 4-[[4′-(N,N-dimethylamino)phenyl]diazenyl]benzoic acid (Dabcyl) as a donor–acceptor pair, linked to a specific sequence for GzmB (AAD), with an additional amino acid as the anchoring point (K). The tetrapeptide was synthesized by microwave-assisted solid-phase peptide synthesis (MW-SPPS) and coupled to Dabcyl and Edans at its N- and C-termini, respectively. The obtained probe was purified by semi-preparative HPLC and characterized by NMR, UV/Vis absorption and fluorescence spectroscopy and mass spectrometry.
Article
The production of lentiviral vectors (LVs) pseudotyped with the vesicular stomatitis virus envelope glycoprotein (VSV‐G) is limited by the associated cytotoxicity of the envelope and by the production methods used, such as transient transfection of adherent cell lines. In this study, we established stable suspension producer cell lines for scalable and serum‐free LV production derived from two stable, inducible packaging cell lines, named GPRG and GPRTG. The established polyclonal producer cell lines produce self‐inactivating (SIN) LVs carrying a WAS‐T2A‐GFP construct at an average infectious titer of up to 4.64 × 10 ⁷ TU mL ⁻¹ in a semi‐perfusion process in a shake flask and can be generated in less than two months. The derived monoclonal cell lines are functionally stable in continuous culture and produce an average infectious titer of up to 9.38 × 10 ⁷ TU mL ⁻¹ in a semi‐perfusion shake flask process. The producer clones are able to maintain a productivity of >1 × 10 ⁷ TU mL ⁻¹ day ⁻¹ for up to 29 consecutive days in a non‐optimized 5 L stirred‐tank bioreactor perfusion process, representing a major milestone in the field of LV manufacturing. As the producer cell lines are based on an inducible Tet‐off expression system, the established process allows LV production in the absence of inducers such as antibiotics. The purified LVs efficiently transduce human CD34 ⁺ cells, reducing the LV quantities required for gene and cell therapy applications.
Chapter
The biggest challenge to immune control of HIV infection is the rapid within-host viral evolution, which allows selection of viral variants that escape from T cell and antibody recognition. Thus, it is impossible to clear HIV infection without targeting "immutable" components of the virus. Unlike the adaptive immune system that recognizes cognate epitopes, the CARD8 inflammasome senses the essential enzymatic activity of the HIV-1 protease, which is immutable for the virus. Hence, all subtypes of HIV clinical isolates can be recognized by CARD8. In HIV-infected cells, the viral protease is expressed as a subunit of the viral Gag-Pol polyprotein and remains functionally inactive prior to viral budding. A class of anti-HIV drugs, the non-nucleoside reverse transcriptase inhibitors (NNRTIs), can promote Gag-pol dimerization and subsequent premature intracellular activation of the viral protease. NNRTI treatment triggers CARD8 inflammasome activation, which leads to pyroptosis of HIV-infected CD4+ T cells and macrophages. Targeting the CARD8 inflammasome can be a potent and broadly effective strategy for HIV eradication.
Thesis
Peptidyl-prolyl isomerases (PPIases) are a ubiquitously expressed super family of proteins that catalyze the cis/trans isomerization of prolyl bonds. Proline conformation acts as a regulatory switch during folding, activation and/or degradation of “clients” that include proteins with roles in cancer and neurodegeneration. PPIase inhibitors, therefore, could be important therapeutics. However, the active site of PPIases is shallow and well conserved between members, challenging selective inhibitor design. Despite this, macrocyclic natural products, including FK506, rapamycin and cyclosporin, bind PPIases with nanomolar or better affinity. These natural products possess an unusual “bifunctional” binding mode and bind two separate proteins simultaneously, which is critical for their activity. They exhibit remarkable pharmacological properties, including oral bioavailability, and rapidly accumulate in cells with widespread tissue distribution. Inspired by this mechanism, I synthesized a library of bifunctional molecules that bind both FKBP12 and HIV protease. Like FK506, we envisioned a model where coincident, high-level expression of both targets - as in HIV-infected lymphocytes with high levels of FKBP12 and HIV protease - would drive cyto-selective sequestration. The library possessed varying affinities for each target, retained passive membrane permeability, and had cellular activity. Molecules highly potent for FKBP12 and HIV protease were selectively taken up into relevant cell populations. Treatment with FKBP12 inhibitors limited partitioning of the molecules, while FKBP12 overexpression enhanced it. This suggests that avidity effects drive selective accumulation of bifunctional molecules in cells expressing high levels of both protein partners. We next focused on Pin1, a unique PPIase that binds prolines directly following phosphoserine/threonine residues. The requirement for an electronegative group in the Pin1 active site renders many inhibitors inactive from poor permeability. We hypothesized that the excellent pharmacological properties of FK506 might make it a suitable scaffold for engineering Pin1 inhibitors. However, FK506 has little affinity for Pin1, because it lacks the key electronegative region essential for Pin1 binding. To overcome this, we designed a novel semisynthetic strategy to modify FK506 at a position to improve affinity for Pin1. Installing a sulfamic acid significantly improved the affinity for Pin1 (>100-fold) in vitro. This strategy is designed to yield high-affinity membrane-permeable inhibitors of Pin1.
Article
Full-text available
During human immunodeficiency virus (HIV) infection there is a profound and selective decrease in the CD4+ population of T lymphocytes. The mechanism of this depletion is not understood, as only a small fraction of all CD4+ cells appear to be productively infected with HIV-1 in seropositive individuals. In the present study, crosslinking of bound gp120 on human CD4+ T cells followed by signaling through the T cell receptor for antigen was found to result in activation-dependent cell death by a form of cell suicide termed apoptosis, or programmed cell death. The data indicate that even picomolar concentrations of gp120 prime T cells for activation-induced cell death, suggesting a mechanism for CD4+ T cell depletion in acquired immune deficiency syndrome (AIDS), particularly in the face of concurrent infection and antigenic challenge with other organisms. These results also provide an explanation for the enhancement of infection by certain antibodies against HIV, and for the paradox that HIV appears to cause AIDS after the onset of antiviral immunity.
Article
Full-text available
In immature thymocytes, T cell receptor for antigen (TCR) mobilization leads to an active T cell suicide process, apoptosis, which is involved in the selection of the T cell repertoire. We have proposed that inappropriate induction of such a cell death program in the mature CD4+ T cell population could account for both early qualitative and late quantitative CD4+ T lymphocyte defects of human immunodeficiency virus (HIV)-infected individuals (Ameisen, J.C., and A. Capron. 1991. Immunol. Today. 4:102). Here, we report that the selective failure of CD4+ T cells from 59 clinically asymptomatic HIV-infected individuals to proliferate in vitro to TCR mobilization by major histocompatibility complex class II-dependent superantigens and to pokeweed mitogen (PWM) is due to an active CD4+ T cell death process, with the biochemical and ultrastructural features of apoptosis. Activation-induced cell death occurred only in the CD4+ T cell population from HIV-infected asymptomatic individuals and was not observed in T cells from any of 58 HIV-seronegative controls, including nine patients with other acute or chronic infectious diseases. Activation-induced CD4+ T cell death was prevented by cycloheximide, cyclosporin A, and a CD28 monoclonal antibody (mAb). The CD28 mAb not only prevented apoptosis but also restored T cell proliferation to stimuli, including PWM, superantigens, and the tetanus and influenza recall antigens. These findings may have implications for the understanding of the pathogenesis of acquired immune deficiency syndrome and for the design of specific therapeutic strategies.
Article
Infection with the human immunodeficiency virus (HIV) is associated with a progressive decrease in CD4 T-cell number and a consequent impairment in host immune defenses. Analysis of T cells from patients infected with HIV, or of T cells infected in vitro with HIV, demonstrates a significant fraction of both infected and uninfected cells dying by apoptosis. The many mechanisms that contribute to HIV-associated lymphocyte apoptosis include chronic immunologic activation; gp120/160 ligation of the CD4 receptor; enhanced production of cytotoxic ligands or viral proteins by monocytes, macrophages, B cells, and CD8 T cells from HIV-infected patients that kill uninfected CD4 T cells; and direct infection of target cells by HIV, resulting in apoptosis. Although HIV infection results in T-cell apoptosis, under some circumstances HIV infection of resting T cells or macrophages does not result in apoptosis; this may be a critical step in the development of viral reservoirs. Recent therapies for HIV effectively reduce lymphoid and peripheral T-cell apoptosis, reduce viral replication, and enhance cellular immune competence; however, they do not alter viral reservoirs. Further understanding the regulation of apoptosis in HIV disease is required to develop novel immune-based therapies aimed at modifying HIV-induced apoptosis to the benefit of patients infected with HIV.
Article
This study investigates apoptosis as a mechanism for CD4+ T-cell depletion in human immunodeficiency virus type-1 (HIV-1) infection. Although several recent studies have shown that T cells of HIV-infected individuals show enhanced susceptibility to cell death by apoptosis, the mechanisms responsible for apoptosis are largely unknown. By using a flow cytometric technique and by morphology, we have quantitated the percentage of cells undergoing apoptosis in peripheral blood mononuclear cells (PBMCs) from HIV-seronegative donors and from HIV- infected asymptomatic patients. The PBMCs were cultured without any stimulus or with staphylococcus enterotoxin B, anti-T-cell receptor (TCR) alpha beta monoclonal antibody WT-31, or phytohemagglutinin for periods up to 6 days. In addition, we sought to determine whether cross- linking of CD4 followed by various modes of TCR stimulation in vitro could induce apoptosis in normal PBMCs. Here we show that (1) patient PMBCs undergo marked spontaneous apoptosis; (2) stimulation of T cells of patients as well as normal donors results in increased apoptosis; and (3) cross-linking of CD4 molecules is sufficient to induce apoptosis in CD4+ T cells if cross-linking is performed in unfractioned PBMCs, but not if CD4 molecules are cross-linked in purified T-cell preparations. These observations strongly suggest that accelerated cell death through apoptosis plays an important role in the pathogenesis of HIV-1 infection. At the same time, our observations implicate cross- linking of CD4 in vivo as a major contributor to this mechanism of accelerated cell death in HIV infection.
Article
This study investigates apoptosis as a mechanism for CD4+ T-cell depletion in human immunodeficiency virus type-1 (HIV-1) infection. Although several recent studies have shown that T cells of HIV-infected individuals show enhanced susceptibility to cell death by apoptosis, the mechanisms responsible for apoptosis are largely unknown. By using a flow cytometric technique and by morphology, we have quantitated the percentage of cells undergoing apoptosis in peripheral blood mononuclear cells (PBMCs) from HIV-seronegative donors and from HIV- infected asymptomatic patients. The PBMCs were cultured without any stimulus or with staphylococcus enterotoxin B, anti-T-cell receptor (TCR) alpha beta monoclonal antibody WT-31, or phytohemagglutinin for periods up to 6 days. In addition, we sought to determine whether cross- linking of CD4 followed by various modes of TCR stimulation in vitro could induce apoptosis in normal PBMCs. Here we show that (1) patient PMBCs undergo marked spontaneous apoptosis; (2) stimulation of T cells of patients as well as normal donors results in increased apoptosis; and (3) cross-linking of CD4 molecules is sufficient to induce apoptosis in CD4+ T cells if cross-linking is performed in unfractioned PBMCs, but not if CD4 molecules are cross-linked in purified T-cell preparations. These observations strongly suggest that accelerated cell death through apoptosis plays an important role in the pathogenesis of HIV-1 infection. At the same time, our observations implicate cross- linking of CD4 in vivo as a major contributor to this mechanism of accelerated cell death in HIV infection.
Article
Members of the tumor necrosis factor (TNF) family such as CD95 (APO-1/Fas) ligand (L) trigger apoptosis in lymphoid cells. Recently, a new member of apoptosis-inducing ligands, TRAIL (TNF-related-apoptosis-inducing-ligand)/Apo-2 ligand, was identified that might act in a similar way. We compared TRAIL and CD95L-induced apoptosis in human lymphoid cells. Expression of TRAIL was found in CD4+ and CD8+ T cells following activation, suggesting that TRAIL participates in T cell-mediated induction of apoptosis. Similar to CD95L, TRAIL-induced apoptosis in target cells is mediated by activation of caspases (ICE/Ced-3 proteases). However, different human lymphoid cell lines and peripheral T cells differ in sensitivity towards induction of apoptosis by TRAIL and CD95L. In addition, T cells are highly sensitive towards CD95L-induced apoptosis after prolonged activation in vitro, but remain completely resistant to TRAIL-induced apoptosis. In contrast, T cells from HIV-1-infected patients previously shown to exhibit increased CD95 sensitivity are even more susceptible towards TRAIL-induced cell death. These data suggest that TRAIL might participate in CD95-independent apoptosis of lymphoid cells and might be involved in deregulated apoptosis in diseases such as leukemias and HIV-1 infection.
Article
Apoptosis is a major mechanism whereby HIV-1 depletes uninfected CD4 F and CD8 F T cells. We previously showed that resting peripheral blood T cells derived from healthy donors were killed by an apoptotic mechanism after adsorption to gp120-containing, protease-defective HIV-1 (L-2) particles, more effectively than parental wild-type LAI adsorption or rgp120-mediated CD4 crosslinking, followed by mitogenic stimulation. Here, we present evidence that the L-2 particle-based apoptosis was induced both in CD4 F and CD8 F cells by generation of effector cells which were mainly derived from a resting memory CD4 F CD38 ‐ subset. This subset enhanced the CD25 expression on the surface and secreted IFN-g in the culture supernatant after L-2 particle exposure. Significant elevation of Fas ligand mRNA was found in the subset by L-2 particle exposure, while expression of Fas antigen on uninfected T cells was induced by exposure to IFN-g. These results indicate that L-2 particles can shift the CD4 F CD38 ‐ subpopulation from a resting to an activated state, and this activation leads to killing of bystander CD4 F and CD8 F T cells by a Fas-mediated mechanism. In fact, purified CD4 F CD38 ‐ cells exposed to L-2 particles were converted into effector cells that were able to kill autologous as well as allogenic target T cells pretreated with IFN-g. Further, we found that the observation of apoptosis due to L-2 particles was a more general phenomenon, that also occurred with Thai primary HIV-1 isolates. These results suggest that such specific types of HIV-1 particles may play a major role in the induction of apoptosis for both bystander CD4 F and CD8 F T cells, through inappropriate activation of CD4 F CD38 ‐ cells.
Article
Treatment of cells with the HIV drugs ritonavir, saquinavir, or nelfinavir (Nfv) inhibits apoptosis induced by a variety of stimuli. Because these drugs are protease inhibitors, they have been postulated to inhibit apoptosis by blocking caspase activity. This study shows that Nfv has no effect on caspase activity or on the transcription or synthesis of a variety of apoptosis regulatory molecules. Instead, Nfv inhibits mitochondrial transmembrane potential loss (Δψm) and the subsequent release of apoptotic mediators. Consequently, the antiapoptotic effects of Nfv are restricted to apoptotic pathways that involve Δψm.