Book a Demo!
CoCalc Logo Icon
StoreFeaturesDocsShareSupportNewsAboutPoliciesSign UpSign In
Download
29547 views
1
2
3
4
5
Background
6
Whole-genome expression profiling exemplified by the
7
development of DNA microarrays represents a major advance
8
in genome-wide functional analysis [ 1 2 ] . In a single
9
assay, the transcriptional response of each gene to a
10
change in cellular state can be measured, whether it is a
11
viral infection, host cell cycle changes, chemical
12
treatment, or genetic perturbation. Specifically,
13
systematic approaches for identifying the biological
14
functions of cellular genes altered during these changes,
15
such as HIV-1 infection, are needed to ensure rapid
16
progress in defining significant host and viral genome
17
sequences in directed experimentation and applications.
18
Therefore, host cellular states can be inferred from the
19
expression profiles, and the notion that the global
20
transcriptional response constitutes a detailed molecular
21
phenotype, such as class discovery, class prediction, drug
22
target validation, and the classification of tumors by
23
expression profiling has begun to receive considerable
24
attention [ 3 4 5 6 7 8 9 10 11 ] .
25
Since its discovery, much of the mainstream human
26
immunodeficiency virus type 1 (HIV-1) Tat research has
27
focused on its ability to activate the HIV-1 LTR. However,
28
to date, besides the transactivation activity on the HIV-1
29
promoter, few other effects exerted by HIV-1 Tat on
30
cellular and viral genes has also been observed. The Tat
31
protein has been shown to transcriptionally repress host
32
cellular genes and be involved in the immunosuppression
33
associated with viral infection. For instance, HIV-1
34
infection is able to down-regulate major histocompatibility
35
complex type I (MHC-I) by various different viral proteins,
36
including Tat which represses the transcription of MHC-I,
37
Vpu which retains nascent MHC-I chains in the endoplasmic
38
reticulum, and Nef which can mediate selective
39
internalization of MHC-I molecules from the plasma
40
membrane. MHC class I gene expression has also been shown
41
to be reduced upon infection with the wild-type LAI virus
42
or a Tat exon one recombinant virus [ 12 13 ] .
43
Tat has been shown to down-regulate mannose receptor,
44
EDF-1, CD3-gamma, and TCR/CD3 surface receptor [ 14 ] . Tat
45
reduces mannose receptor levels and promoter activity in
46
mature macrophages and dendritic cells by interfering with
47
the host transcriptional machinery; resulting in decreased
48
levels of surface mannose receptor needed for Ag
49
(mannosylated albumin uptake) or pathogen capture
50
(Pneumocystis carinii phagocytosis), and eventual delivery
51
to MHC class II-containing intracellular compartments [ 15
52
] . EDF-1, a gene down-regulated when endothelial cells are
53
induced to differentiate
54
in vitro , was shown to be
55
down-regulated by Tat at the transcriptional level,
56
resulting in the inhibition of endothelial cell growth and
57
in the transition from a nonpolar cobblestone phenotype to
58
a polar fibroblast-like phenotype [ 16 ] .
59
When examining the
60
in vivo effects of HIV-1 Tat protein
61
in the
62
Xenopus embryo, it was found that
63
upon injection of synthetic Tat mRNA into zygotes, a marked
64
delay in gastrulation occurred. This led to the altered
65
specification of the anterior-posterior axis and partial
66
loss of the anterior embryo structures. Mechanistically,
67
HIV-1 Tat elicited a general suppression of gene
68
expression, including that of
69
Xbra and
70
gsc , two early genes whose
71
expression are required for proper gastrulation [ 17 ]
72
.
73
In relation to the cell cycle, Tat has also been shown
74
to bind to p53 and inhibit the transcription of p53
75
responsive elements, such as the p21/Waf1 gene promoter.
76
Consequently, upon introduction of stress signals (e.g.,
77
gamma irradiation), HIV-1-infected cells lose their G1/S
78
checkpoint, enter the S-phase inappropriately, and apoptose
79
[ 18 19 20 ] . Finally, the inhibition of Tat on
80
translational machinery has also been noted. The potential
81
translational inhibitory effects of the TAR RNA region is
82
mediated by the activation of p68 (the interferon-induced
83
68-kilodalton protein kinase) kinase, which was
84
down-regulated by Tat during productive HIV-1 infection [
85
22 ] .
86
Although the mechanism of the host cellular
87
down-regulation remains largely unknown, few reports have
88
attempted to decipher the mechanism of the observed
89
inhibition. For instance, the addition of Tat to PC12 cells
90
up-regulated the expression of the inducible cAMP early
91
repressor (ICER), a specific member of the cAMP-responsive
92
element modulator transcription factor family, in a
93
cAMP-dependent manner. In turn, ICER overexpression
94
abrogated the transcriptional activity of the TH promoter,
95
strongly suggesting ICER's involvement in Tat-mediated
96
inhibition of gene expression [ 23 ] .
97
Aside from induction of ICER, Tat is capable of forming
98
complex (es) with a component of TFIID, TAF
99
II 250 [ 24 ] and Tip60 [ 25 ] both of
100
which contain histone acetyltransferase (HAT) activity. In
101
these cases, Tat-TAF
102
II 250 and Tat-Tip60 do not affect the
103
transcription from the HIV-1 LTR, but interfere with the
104
transcription activity of cellular genes. It is postulated
105
that different targets of HATs by Tat have different
106
consequences. The interaction of Tat with p300/CBP and
107
P/CAF stimulates its ability to transactivate LTR-dependent
108
transcription, while Tat-TAF
109
II 250 or Tat-Tip60 interactions control
110
the transcription of cellular genes.
111
Here to better understand the host response to Tat, we
112
have performed microarray experiments on HIV-1 infected
113
cells expressing the Tat protein. To our surprise many host
114
cellular genes were down-regulated when comparing HIV-1
115
infected latent cells to uninfected parental cells. Because
116
most, if not all, latent infected cells available to date
117
(e.g., ACH2, U1, J1.1, OM.10) have various expression
118
levels of doubly spliced viral mRNAs, including Tat, Rev,
119
Nef, Vpr, and other accessory proteins, we decided to
120
perform the microarray in a system where Tat was
121
constitutively expressed; asking whether Tat by itself, or
122
in the absence of other accessory proteins, could still
123
down-regulate host cellular genes. Consistent with latently
124
infected cells, we found many cellular genes to be
125
down-regulated in Tat expressing lymphocytes. The
126
down-regulation is most apparent on cellular receptors that
127
have intrinsic receptor tyrosine kinase (RTK) activity and
128
signal transduction members that mediate RTK function;
129
including the Ras-Raf-MEK pathway, and co-activators such
130
as p300/CBP and SRC-1, which mediate gene expression
131
related to hormone receptor genes. Interestingly, we also
132
observed up-regulation of S-phase genes, as well as
133
ribosomal genes involved in translation. Functionally,
134
down-regulation of receptors may allow latent HIV-1
135
infected cells to either hide from the immune system or
136
avoid extracellular differentiation signals normally
137
regulated by receptors. Up-regulation of S-phase and
138
translation genes may allow speeding of cells through the
139
S-phase and subsequent accumulation at the G2 phase, where
140
most of the cellular and viral translation may take place.
141
Therefore, the presence of Tat may not only control
142
activated transcription on HIV-1 LTR, but also aid in the
143
subsequent translation of viral mRNA in the cytoplasm.
144
145
146
Results and discussion
147
148
Receptor family members
149
It has long been known that infection by HIV-1
150
commonly leads to the down-regulation and the
151
disappearance of CD4 receptors from the plasma membrane,
152
a phenomenon referred to as receptor down-modulation.
153
This, in turn, renders cells refractory to subsequent
154
infection by the same or other viruses that use the CD4
155
receptor for entry; thus creating a state of
156
super-infection immunity. Results in Table 1indicate that
157
although few receptor genes were up-regulated, most of
158
the cellular receptors in general, were down-regulated in
159
the presence of the Tat protein. Most of these receptors
160
or membranous proteins were initially discovered from
161
immune or neuronal cells, hence they were given names
162
related to the immune or nervous system. For instance,
163
mRNA for the neuropeptide Y-like receptor (Acc# X71635),
164
which was up-regulated in Tat expressing cells, was
165
initially discovered as a G-protein coupled neuropeptide
166
Y receptor, and later found to be homologous to the
167
co-receptor CCR5 needed for HIV-1 infection of
168
monocyte/macrophage cells. Therefore, most of the
169
receptors listed in Table 1may in fact be expressed in
170
various tissues and have multiple functions.
171
Consistent with our microarray results on CCR5
172
up-regulation, experiments performed in peripheral blood
173
mononuclear cells (PBMCs) with soluble Tat has shown
174
selective entry and replication of CCR5 virus into cells
175
[ 27 28 ] . Up-regulation of HIV-1 coreceptor by Tat has
176
also been reported, where a synthetic Tat protein that
177
was immobilized on a solid substrate, up-regulated the
178
surface expression of the chemokine receptors in purified
179
populations of primary resting CD4+ T cells. Also, a
180
similar result was seen from Tat protein actively
181
released by HIV-1 infected cells, implying a potentially
182
important role for extracellular Tat in rendering the
183
bystander CD4+ T cells more susceptible to infection [ 28
184
] .
185
We therefore tested whether H9/Tat cells, which showed
186
an increase in CCR5 expression, could in fact allow
187
better entry and infection of the CCR5 (R5) virus into
188
cells. Figure 3Ashows the result of such an experiment,
189
where H9/Tat cells allowed a better replication profile
190
of the R5 than the CXCR4 (X4) virus. The increase in
191
viral titer peaked after some 18 days of infection with
192
the R5 virus, further implying that the CCR5 co-receptor
193
allowed a better selection of R5 virus in Tat expressing
194
cells.
195
Another example of co-receptors with multiple
196
functions is the leukotriene family member B4, which was
197
down-regulated in Tat expressing cells (Acc# D89078,
198
Table 1). The cysteinyl leukotrienes (CysLT), LTC, LTD,
199
and LTE, were first shown to be essential mediators in
200
asthma [ 29 ] . However, when the mouse leukotriene B4
201
receptor (m-BLTR) gene, was cloned it was shown to have
202
significant sequence homology with chemokine receptors
203
(CCR5 and CXCR4), co-receptors for many different HIV-1
204
clades [ 30 ] . Along the same lines, when cells were
205
infected with 10 primary clinical isolates of HIV-1,
206
leukotriene B4 receptor was primarily utilized for
207
efficient entry into cells which were mainly of the
208
syncytium-inducing phenotype [ 31 ] . Therefore,
209
up-regulation of neuropeptide Y-like receptor and
210
down-regulation of leukotriene B4 receptor in Tat
211
expressing cells indicates a selective advantage of one
212
class of virus (CCR5) over another (CXCR4).
213
Other examples of consistency between our microarray
214
results on receptors and the HIV-1 Tat literature,
215
include the down-regulation of gene expression in uPAR
216
(Acc# X74039), IP3 (Acc# D26070, D26351), Glu R flop
217
(Acc# U10302), PPAR (Acc# L07592), alpha-2 macroglobulin
218
receptor protein (Acc# M63959), and receptor tyrosine
219
kinase (Acc# L36645, U66406) genes.
220
The transmembranous urokinase-type plasminogen
221
activator receptor (uPAR; CD87) focuses the formation of
222
active plasmin at the cell surface, thus enhancing
223
directional extracellular proteolysis. Interestingly, the
224
promoter activity of the CD87 gene was shown to decline
225
after infection [ 32 ] , implying that post integration
226
of HIV-1 may in fact down-regulate CD87 gene expression.
227
Similarly, inositol 1,4,5-trisphosphate receptors (IP3R)
228
are intracellular calcium release channels involved in
229
diverse signaling pathways and are required for the
230
activation of T lymphocytes [ 33 ] . Tat (also implicated
231
as a neurotoxin) has been shown to release calcium from
232
inositol 1,4, 5-trisphosphate (IP3) receptor-regulated
233
stores in neurons and astrocytes causing premature
234
apoptosis [ 34 ] . Down-regulation of IP3 may therefore
235
contribute to viral latency and maintenance of an
236
anti-apoptotic state in cells.
237
HIV-1 infection can cause extensive neuronal loss and
238
clinically, a severe dementia. The cause of the
239
neurotoxicity remains unclear as neurons are not
240
infected, but the disturbance of glutamate-linked calcium
241
entry has been implicated. It has been shown that
242
HIV-infected brain has a decrease of mRNA and protein of
243
the GluR-A flop subtype of
244
alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
245
(AMPA) glutamate receptor in cerebellar Purkinje cells.
246
The observed disturbance of AMPA receptors may contribute
247
to the neurotoxic process in other vulnerable brain
248
regions and clinically to the development of dementia [
249
35 ] . Interestingly, in a mouse model AMPA receptors in
250
the cortex, striatum, hippocampus, and cerebellum
251
declined by 29-50% as early as 8 weeks post-retroviral
252
inoculation. Thus, the reduction in AMPA receptor density
253
may contribute to the development of the cognitive
254
abnormalities associated with HIV-1 infection [ 36 ]
255
.
256
Finally, patients with AIDS who are receiving therapy
257
with HIV-1 protease inhibitors have been reported to be
258
afflicted with a syndrome characterized by lipodystrophy
259
(fat redistribution favoring the accumulation of
260
abdominal and cervical adipose tissue), hyperlipidemia,
261
and insulin resistance. Potential mechanisms for altered
262
adipocyte function include, direct binding to PPARgamma
263
or inhibition of transcription of PPARgamma promoter [ 37
264
] . The lipodystrophy syndrome may be a result of the
265
inhibition of 2 proteins involved in lipid metabolism
266
that have significant homology to the catalytic site of
267
HIV proteases; namely cytoplasmic retinoic acid binding
268
protein type 1 and low density
269
lipoprotein-receptor-related protein [ 38 ] . An
270
additional mechanism of PPAR down-regulation may be
271
related to Tat expression in latent cells.
272
273
274
Translation associated factors
275
Viruses have evolved a remarkable variety of
276
strategies to modulate the host cell translation
277
apparatus with the aim of optimizing viral mRNA
278
translation and replication. For instance, viruses
279
including Herpes simplex virus type 1 (HSV-1) have been
280
known to induce severe alterations of the host
281
translational apparatus, including the up-regulation of
282
ribosomal proteins and the progressive association of
283
several nonribosomal proteins, such as VP19C, VP26, and
284
the poly(A)-binding protein 1 (PAB1P) to ribosomes [ 39 ]
285
. In the case of HIV-1, approximately one infectious
286
HIV-1 genome in an infected cell could be transcribed and
287
translated into 50,000 to 100,000 physical particles [ 40
288
] . This poses an immense challenge for the virus to be
289
able to transcribe, splice, transport, and translate its
290
RNA into fully packaged virions in a timely fashion.
291
Therefore, it would be advantageous for the virus to set
292
the stage for each successive step necessary for viral
293
progeny formation. One such event is Tat's ability to
294
control genes that aid in translational machinery. As
295
seen in Table 2, many of the critical components of a
296
functional ribosome, including large subunits L 3, 6, 26,
297
31, 38, and 41, as well as S 6, 12, 20, and 24, and many
298
of the translation initiation factors are up-regulated by
299
Tat. This would imply that Tat up-regulates many
300
ribosomal genes that may be necessary to produce
301
functional ribosomes needed for viral mRNA translation.
302
Therefore, interfering with translation could provide a
303
new strategy for anti-HIV treatment. Along these lines,
304
when the aminogylcosides (kanamycin, hygromycin B,
305
paromycin and neomycin) due to their ability to inhibit
306
protein synthesis by affecting ribosomal fidelity, or
307
puromycin because of its competing ability with tRNAs for
308
binding on the large ribosomal subunit, or cycloheximide
309
which inhibit the large ribosomal subunit by preventing
310
ribosomal movement along the mRNA, were used in active
311
HIV-1 infection, it was found that both cycloheximide and
312
puromycin produced the greatest decrease in HIV-1
313
inhibition, presumably by inhibiting the large subunit of
314
the ribosome [ 41 ] .
315
Translation of HIV-1 RNAs pose a challenge since they
316
all contain a TAR sequence at their 5' end. The
317
Tat-responsive region (TAR) of HIV-1 exhibits a
318
trans-inhibitory effect on translation by activating the
319
interferon-induced 68-kilodalton protein kinase.
320
Productive infection by HIV-1 has been shown to result in
321
a significant decrease in the amount of cellular p68
322
kinase. The steady-state amount of p68 kinase was found
323
to be reduced in cells stably expressing Tat. Thus, the
324
potential translational inhibitory effects of the TAR RNA
325
region, mediated by activation of p68 kinase, may be
326
down-regulated by Tat during activation of the latent
327
virus [ 22 ] . Along these lines, a Tat peptide
328
antagonist, which bound specifically to TAR RNA and
329
competed with Tat for binding, reduced Tat-dependent
330
translation [ 42 ] .
331
Finally, upregulation of translation genes in Tat
332
expressing cells is specially intriguing in light of the
333
recent discovery of internal ribosome entry sites (IRESs)
334
in HIV-1 gag ORF [ 43 ] . IRESs are thought to promote
335
initiation of translation by directly binding to
336
ribosomes, in a manner independent of the mRNA cap or of
337
scanning through upstream sequences. Since, the TAR is
338
located at the 5' end of all HIV-1 RNA transcripts and
339
the presence of secondary structure at or near the 5' end
340
of RNAs reduces the accessibility of the 5' cap to eIF4F,
341
it is thought that this feature of HIV-1 mRNAs can
342
inhibit their cap-dependent translation [ 44 45 46 ] .
343
Therefore, a possible function of the HIV-1 gag IRES
344
might be to serve as a mechanism to bypass the structural
345
barriers to cap-dependent translation by recruiting
346
ribosomes easily and directly to the gag ORFs. IRES
347
entirely contained within a translated ORF has been shown
348
in the MMLV gag [ 47 ] , and host mRNA encoding p110
349
PITSLREand p58 PITSLRE [ 48 ] . Along these lines,
350
cap-dependent translation may be cell cycle regulated,
351
especially when cells are arrested at the G2 phase of the
352
cell cycle, where the cap-dependent translation of most
353
cellular host cell mRNAs is inhibited [ 49 50 51 ] .
354
355
356
Modulation of signal transduction pathway
357
Results in Table 3indicate that many seemingly
358
different pathways are being regulated by Tat. However,
359
the signal transduction pathway, MAPK, has been shown to
360
control and be upstream of DNA-replication,
361
transcription, and cell cycle pathways [ 52 53 54 ] . The
362
mitogen-activated protein kinase (MAPK) pathway,
363
consisting of the MAP kinase kinases (MKKs) 1 and 2, and
364
extracellular signal-regulated kinases (ERKs) 1 and 2,
365
which have been implicated in diverse cellular processes
366
including proliferation, transformation, and cell
367
differentiation [ 53 ] . The MAP kinase (MAPK) pathway
368
has emerged as a crucial route between membrane-bound Ras
369
and the nucleus. This MAPK pathway encompasses a cascade
370
of phosphorylation events involving three key kinases,
371
namely Raf, MEK (MAP kinase kinase) and ERK (MAP kinase).
372
The MAPK pathway controls ERKs 1 and 2, c-Jun N-terminal
373
kinase (JNK), and p38. These signaling pathways in turn,
374
activate a variety of transcription factors including
375
NF-kappaB (p50/p65), AP-1 (c-Fos/c-Jun), and CREB
376
phosphorylation, which in turn coordinate the induction
377
of many genes encoding inflammatory mediators.
378
Cytokine receptors such as IL-3, GM-CSF, and the
379
interferons transmit their regulatory signals primarily
380
by the receptor-associated Jak family of tyrosine
381
kinases, and activate STAT transcription factors.
382
Activated STAT5 proteins are detected in reduced levels
383
in lymphocytes recovered from HIV-infected patients and
384
immunocompromised mice. Both of these types of receptor
385
signaling pathways have recently been shown to interact
386
with serine/threonine kinases such as MAP kinases. A
387
common intermediate pathway initiating from receptors to
388
the nucleus is the Ras/Raf/MEK/ERK (MAPK) cascade, which
389
can result in the phosphorylation and activation of
390
additional downstream kinases and transcription factors
391
such as p90Rsk, CREB, Elk, and Egr-1 [ 55 56 ] .
392
Therefore, it is intriguing that Tat expressing cells
393
show down-regulation of MAPK components (Table 3, Figure
394
3Band 3C), essential mediators between receptors and
395
nuclear transcription factors. This would imply that
396
latently infected cells that express Tat (doubly spliced
397
RNA) and not the whole virus (all three classes of the
398
RNA), can control signal transduction related to membrane
399
and transcriptional signaling (Figure 6).
400
Interestingly, Tat, through the RGD motif, which
401
controls integrin-based cell signaling, has been reported
402
to mediate the activity of phosphotyrosine
403
phosphatase(s). This in turn which would lead to a
404
decrease in the levels of phosphotyrosine-containing
405
proteins such as ERK-2/p42MAPK kinases [ 57 ] .
406
Cysteine-rich and basic Tat peptides have been shown to
407
inhibit VEGF-induced ERK activation and mitogenesis.
408
These peptides also inhibited proliferation,
409
angiogenesis, and ERK activation induced by basic
410
fibroblast growth factor with similar potency and
411
efficacy [ 58 ] . Consistent with this model, it has been
412
shown that treatment of neural cells with culture
413
supernatants from HAART-treated subjects, which
414
presumably contain extracellular Tat, resulted in
415
down-regulation of the JNK, AKT, and ERK kinases [ 59 ]
416
.
417
Finally, activation of MAPKs has been shown to
418
activate the singly spliced and unspliced (genomic)
419
latent HIV-1 virus. For instance, the signal transduction
420
pathways that regulate the switch from latent to
421
productive infection have been linked to MAPK. The
422
induction of latent HIV-1 expression has been shown to be
423
inhibited by PD98059 and U0126, specific inhibitors of
424
MAPK activation. The MAPK acts by stimulating AP-1 and a
425
subsequent physical and functional interaction of AP-1
426
with NF-κB, resulting in a complex that synergistically
427
transactivates the HIV-1 [ 60 ] . At the level of
428
infection and entry, the activation of MAPK through the
429
Ras/Raf/MEK (MAPK kinase) signaling pathway enhances the
430
infectivity of HIV-1 virions. Virus infectivity can be
431
enhanced by treatment of cells with MAPK stimulators,
432
such as serum and phorbol myristate acetate, as well as
433
by coexpression of constitutively activated Ras, Raf, or
434
MEK in the absence of extracellular stimulation [ 61 ] .
435
Also, following infection, efficient disengagement of the
436
reverse transcription complex from the cell membrane and
437
subsequent nuclear translocation, requires
438
phosphorylation of the reverse transcription complex
439
components by ERK/MAPK; demonstrating a critical
440
regulation of an early step in HIV-1 infection by the
441
host cell MAPK signal transduction pathway [ 62 ] .
442
Therefore, Tat down-regulation of the MAPK pathway in
443
latent cells implies that much of the host signal
444
transductions connected to activation are down-regulated,
445
and at the same time, these cells may be refractory to
446
subsequent infection by other viruses.
447
448
449
Thymosin family members, and cell cycle
450
Prothymosin α (ProTα) belongs to the α-Thymosin family
451
which comprises different polypeptides widely distributed
452
within animal tissues. Although its role has remained
453
controversial, it is involved in the increase of
454
immediate early genes such as c-myc [ 63 ] , which is
455
upstream of cyclin D synthesis and necessary for cell
456
division [ 64 ] . In humans, ProTα is coded by a gene
457
family of six members. One of them contains introns,
458
exons and classic regulatory signals, while the remaining
459
five are intronless genes [ 59 ] located on chromosome 2
460
[ 66 ] . There are two mRNA transcripts, which arise in a
461
ratio of 9:1 (shorter/longer form), where only the long
462
transcript is regulated by extracellular signals.
463
It has been demonstrated that malignant tissues with
464
accelerated cell cycle show higher levels of ProTα
465
expression than normal or surrounding healthy tissues [
466
67 ] . ProTα was shown as a marker for breast cancer [ 68
467
] , hepatocarcinoma [ 69 ] , and plasma levels of its
468
derivative Tα
469
1 been proposed as a marker for the
470
prognosis of lung cancer [ 70 ] . In ligand blotting
471
assays, ProTα bound only to chromatin pools and nuclear
472
fractions where histone H1 was present [ 71 72 ] . The
473
analysis of the interaction of ProTα with H1-containing
474
chromatin suggests a putative role for ProTα in the
475
fine-tuning of the stoichiometry and/or mode of
476
interaction of H1 with chromatin [ 73 ] . Interestingly,
477
HL-60 cells overexpressing ProTα show an enhancement of
478
accessibility of micrococcal nuclease to chromatin,
479
implying relaxed chromatin structure for enhanced cell
480
cycle gene expression [ 74 ] .
481
A broad study using several mononuclear and
482
fibroblastic cell lines has shown that ProTα mRNA
483
accumulation is cell cycle phase-dependent. In the U937
484
monocytic cell line, ProTα mRNA peaked at the end of S/G2
485
phase and fell towards the entry into the new G1 phase.
486
More prominent mRNA regulation was found in the
487
fibroblastic cell lines CV1 and NIH3T3, with peak mRNA
488
levels at the end of S-phase. In all cases the expression
489
pattern coincided with that of cyclin B and Cdc2/cyclin B
490
activation [ 75 ] .
491
It is interesting to note that Cdc2 (Acc# X05360),
492
Cdc10 homolog (Acc# S72008), and Cdc37 (Acc# U43077) were
493
all up-regulated in Tat expressing cells. Cdc2, a
494
catalytic subunit of cyclin-dependent kinases, is
495
required for both the G1-to-S and G2-to-M transitions. In
496
the fission yeast Schizosaccharomyces pombe, the
497
execution of Start requires the activity of the Cdc2
498
protein kinase and the Cdc10/Sct1 transcription complex.
499
The loss of any of these genes leads to G1 arrest [ 69 ]
500
.
501
Cdc37 encodes a 50-kDa protein that targets
502
intrinsically unstable oncoprotein kinases including
503
Cdk4, Raf-1, and v-src to the molecular chaperone Hsp90,
504
an interaction that is thought to be important for the
505
establishment of signaling pathways. Cdc37 expression may
506
not only be required to support proliferation in cells
507
that are developmentally programmed to proliferate, but
508
may also be required in cells that are inappropriately
509
induced to initiate proliferation by oncogenes. For
510
instance, MMTV-Cdc37 transgenic mice develop mammary
511
gland tumors at a rate comparable to that observed
512
previously in MMTV-cyclin D1 mice, indicating that Cdc37
513
can function as an oncogene in mice and suggests that the
514
establishment of protein kinase pathways mediated by
515
Cdc37-Hsp90 can be a rate-limiting event in
516
transformation [ 76 ] . Also, analysis of proteins that
517
co-immunoprecipitated with Cdk6 and Cdk4 has shown
518
complexes containing both Hsp90 and Cdc37 [ 77 78 79 ]
519
.
520
Cdc37 also promotes the production of Cak1. Cak1 in
521
yeast is the human homolog of CAK trimeric enzyme
522
containing CDK7, cyclin H, and MAT1. Both human and yeast
523
Caks function as RNA polymerase II CTD kinase, Cdk
524
activating kinase, and DNA damage/repair enzymes. Cdc37,
525
like its higher eukaryotic homologs, promotes the
526
physical integrity of multiple protein kinases, perhaps
527
by virtue of a cotranslational role in protein folding [
528
80 ] . Finally, Hsp90/Cdc37 has recently been shown in
529
the stabilization/folding of Cdk9 as well as the assembly
530
of an active Cdk9/cyclin T1 complex responsible for
531
P-TEFb-mediated Tat transactivation [ 81 ] .
532
533
534
Transcription and chromatin remodeling
535
factors
536
A highly ordered chromatin structure presents a
537
physical obstacle for gene transcription; presumably by
538
limiting the access of transcription factors and RNA
539
polymerase II core machinery to target DNA [ 82 83 ] . In
540
concert with the observation that corepressors are
541
associated with HDAC activities [ 84 85 ] , it appears
542
that the transcriptional outcome of nuclear receptors is
543
determined by the balance of histone acetylation and
544
deacetylation activities, and that ligands serve as a
545
switch to recruit HATs with the concomitant dismissal of
546
HDACs. Signal transduction pathways add another layer of
547
regulation to the functions of CBP/p300. In the case of
548
the POU homeodomain factor Pit-1, transcriptional
549
activity is potentiated by MAPK pathways [ 86 ] .
550
Therefore, down-regulation of MAPK pathway members in Tat
551
expressing cells, as seen in Table 3, is consistent with
552
decreased phosphorylation of DNA binding factors such as
553
Pit-1, and overall lower DNA binding activity. Here, we
554
describe the effect of coactivator proteins SRC-1 (Acc#
555
AJ000882, U90661, Table 1) and p300 (Acc# U01877, Table
556
3), and their relation to differentiation genes such as
557
retinoic acid receptor (RAR/PML, Acc#: X06614, Table 1),
558
and Leptin receptor variant (Acc#: U66496, Table 1); all
559
of which are down-regulated in Tat expressing cells
560
(Figure 5).
561
Over the past three decades a great deal of evidence
562
has accumulated in favor of the hypothesis that steroid
563
receptor hormones act via regulation of gene expression.
564
The action is mediated by specific nuclear receptor
565
proteins, which belong to a superfamily of
566
ligand-modulated transcription factors that regulate
567
homeostasis, reproduction, development, and
568
differentiation [ 87 ] . This family includes receptors
569
for steroid hormones, thyroid hormones, hormonal forms of
570
vitamin A and D, peroxisomal activators, and ecdysone [
571
88 ] . Nuclear hormone receptors are ligand-dependent
572
transcription factors that regulate genes critical to
573
such biological processes as development, reproduction,
574
and homeostasis. Interestingly, these receptors can
575
function as molecular switches, alternating between
576
states of transcriptional repression and activation,
577
depending on the absence or presence of a cognate
578
hormone, respectively. In the absence of cognate hormone,
579
several nuclear receptors actively repress transcription
580
of target genes via interactions with the nuclear
581
receptor corepressors SMRT and NCoR. Upon binding of the
582
hormone, these corepressors dissociate from the DNA-bound
583
receptor, which subsequently recruits a nuclear receptor
584
coactivator (NCoA) complex. Prominent among these
585
coactivators is the SRC (steroid receptor coactivator)
586
family, which consists of SRC-1, TIF2/GRIP1, and
587
RAC3/ACTR/pCIP/AIB-1. These cofactors interact with
588
nuclear receptors in a ligand-dependent manner and
589
enhance transcriptional activation via histone
590
acetylation/methylation and recruitment of additional
591
cofactors such as CBP/p300 [ 89 ] . CBP/p300 has been
592
implicated in the functions of a large number of
593
regulated transcription factors based primarily on
594
physical interaction and the ability to potentiate
595
transcription when overexpressed [ 90 ] . In the case of
596
nuclear receptors, the interaction with CBP/p300 is
597
ligand-dependent and relies on the conserved nuclear
598
receptor functional domain, AF-2 (activation function 2).
599
600
In vivo studies have supported the
601
conclusion that CBP/p300 are components of the
602
hormonal-regulation of transcription in fibroblasts
603
isolated from a p300-/- mouse; and loss of the p300 gene
604
severely affects retinoic acid (RA)-dependent
605
transcription [ 91 ] . In a separate study using
606
hammerhead ribozymes that specifically cleave CBP or p300
607
mRNA, Kawasaki et al [ 92 ] reported that reduced
608
cellular CBP or p300 levels resulted in compromised
609
expression of endogenous RA-inducible genes such as
610
p21/Waf1 and p27 cdk inhibitors. Along this line, Tat
611
expressing cells have lower levels of p21/Waf1 presumably
612
due to inactivation of p53 and a lack of p300/RA- induced
613
gene expression. Consistent with this interpretation, CBP
614
and p300 harbor transcriptional activation of
615
ligand-induced RA or ER function on a chromatinized
616
template [ 93 ] .
617
The NcoA family members constitute SRC-1/NcoA-1 [ 89 ]
618
, TIF2/GRIP1/NcoA-2, [ 94 95 ] and pCIP/ACTR/AIB1 [ 96 97
619
98 ] proteins, which interact with liganded RA receptor
620
(RAR), and CBP/p300. Overexpression of these NCoA factors
621
enhances ligand-induced transactivation of several
622
nuclear receptors [ 99 ] . A weak intrinsic HAT activity
623
has been reported in SRC-1/NCoA-1 and pCIP/ACTR/AIB1,
624
suggesting that chromatin remodeling may also be a
625
function of these NCoA factors [ 99 100 ] ; although they
626
do not appear to contain regions homologous to the HAT
627
domains of CBP/p300 or p/CAF. Structure-function analysis
628
of the NCoAs have revealed multiple copies of a signature
629
motif, LXXLL, with conserved spacing that is required for
630
interaction with nuclear receptors and CBP/p300 [ 99 101
631
] . Intriguingly, different LXXLL motifs are required for
632
PPARγ (Peroxisome Proliferator activated receptor γ, a
633
gene down-regulated in Tat expressing cells; Acc# L07592,
634
Table 1) function in response to different classes of
635
ligands, suggesting distinct configuration of assembled
636
complexes.
637
Taken together, through the use of microarray
638
technology, we have described one of the first
639
observations about how Tat is able to control various
640
host cellular machineries. Although our data is
641
consistent with most of the cited literature on the
642
effects of Tat in infected host and uninfected bystander
643
cells, we caution that the transcriptional profiling in
644
chronically infected cells such as ACH2 or H9/Tat cells
645
may not necessarily be representative of the pattern of
646
expression observed in most cells infected by other group
647
M, N, or O HIV-1 isolates.
648
We recently extended our observations by utilizing
649
other HIV-1 infected cells which normally express Tat
650
(U1), and addition of exogenous purified Tat to
651
uninfected PBMCs. Preliminary results using western blots
652
supports the idea that genes which were altered in H9/Tat
653
system also showed a similar level of change in few of
654
the tested genes (Figure 3C). This notion of consistency
655
was further confirmed using the IL-8 activation by Tat.
656
Interleukin-8 (IL-8) belongs to the CXC chemokine family
657
and is secreted by several different cell types,
658
including monocytes, neutrophils, endothelial cells,
659
fibroblasts, and T lymphocytes. IL-8 production (induced
660
by several stimuli, including IL-1, TNF-, and phorbol
661
myristate acetate) is primarily regulated at the
662
transcriptional level. IL-8 is a potent chemotactic
663
factor for granulocytes and T lymphocytes, and is found
664
in HIV-infected individuals. The CXC chemokine IL-8 does
665
not bind to CCR5. It has previously been shown that IL-8
666
mRNA induction was seen less then 1 h after Tat (72aa)
667
stimulation, and levels remained elevated for up to 24 h,
668
leading to IL-8 protein production [ 102 ] . Along these
669
lines, we have previously shown that the IL-8 gene is
670
expressed in a cell cycle-dependent manner in cells that
671
express the Tat protein, and the induction is during the
672
S phase of the cell cycle and regulated by stable NF-kB
673
binding to the IL-8 promoter [ 103 ] . When looking for
674
IL-8 at the G1/S border, we found that all Tat containing
675
cells, including PBMCs that were treated with exogenous
676
Tat showed an up-regulation of IL-8 in the supernatant
677
(Figure 4), further implying that results obtained from
678
the H9/Tat system may infact be of general physiological
679
relevance in vivo.
680
Finally, throughout the current study we came across
681
some technical findings that were critical in the
682
confirmation of most of our results. For instance, few
683
genes did not correlate in their activation or
684
suppression levels when comparing fold changes between
685
microarrays and protein levels using western blot
686
analysis. We suspect this is because many genes that are
687
transcribed may not necessarily be translated, due to
688
their cell cycle stage, 5' stem and loop RNA structures,
689
varying half-lives of proteins and mRNAs, and a host of
690
other unknown variables. Also, specific changes that
691
occur in a cell may not be required in redundant pathways
692
that score for a specific function. This is commonly seen
693
in the differences between HIV-1 infected or Tat
694
expressing
695
in vitro cell lines and AIDS
696
patients PBMC samples. Therefore, other microarrays would
697
have to be performed on purified infected PBMCs to
698
confirm most of the changes observed in Tables 1, 2, and
699
3. Unfortunately, to date this particular issue is not
700
feasibly addressable, since it is not possible to isolate
701
a homogenous population of infected T- or Monocytic cells
702
from AIDS patients. Also, confirmatory tests for protein
703
expression would have to be done with both hydrophilic
704
and hydrophobic extraction buffers. For instance, we have
705
observed that PCNA protein, which is up-regulated in Tat
706
expressing cells, extract best with hydrophobic buffers
707
from the nucleus, presumably due to its binding to DNA
708
replication machinery (data not shown). Future
709
experiments will address issues related to differences
710
between various HIV-1 Tat clades, host expression levels
711
between T- and Monocytic cells, and its effect at various
712
stages of the cell cycle.
713
714
715
716
Conclusions
717
Expression profiling from HIV-1 or Tat expressing cells
718
holds great promise for rapid functional analysis. Here, we
719
have described the effect of Tat and its alterations with
720
the host cellular gene expression. We observed that more
721
than 2/3 of the cellular genes tested were down-regulated
722
by Tat. These genes belong to receptor, co-receptor, and
723
co-activator pathways that are part of serine/threonine
724
receptor tyrosine kinase, Ras/Raf/MEK/ERK (MAPK) cascade,
725
which control proliferative and/or differentiation signals.
726
We also observed a great deal of increase in the host cell
727
translation apparatus with the possible aim of optimizing
728
viral mRNA translation prior to viral maturation and
729
release. Therefore, HIV-1 accessory doubly spliced messages
730
such as Tat, may control the host gene expression in
731
latently infected cells, and determine not only viral
732
transcription, but also the fate of post-transcriptional
733
events.
734
735
736
Materials and method
737
738
Cell culture
739
ACH
740
2 cells are HIV-1 infected CD4
741
lymphocytic cells, with an integrated wild-type
742
single-copy chromatinized DNA. The CEM T cell (12D7) is
743
the parental cell for ACH
744
2 cells. ACH2 cell lines has a single
745
copy of LAI strain proviral sequence. The TAR has a point
746
mutation at (C37 -> T), which no longer responds
747
(efficiently) to Tat. However, the cell line is fully
748
capable of making infectious virus in presence of TNF,
749
PHA, PMA, and a host of other stimuli. H9 and H9/Tat
750
cells are both CD4+ Lymphocytic cells, where H9 cells
751
carry a control integrated vector without the Tat open
752
reading frame, and H9/Tat cells carry integrated Tat
753
expression vector. Both cell lines were a generous gift
754
of George Pavlakis (NCI, NIH). U1 is a monocytic clone
755
harboring two copies of the viral genome from parental
756
U973 cells. All cells were cultured at 37°C up to 10
757
5cells per ml in RPMI-1640 media, containing 10% Fetal
758
Bovine Serum (FBS) treated with a mixture of 1%
759
streptomycin and penicillin antibiotics, and 1%
760
L-glutamine (Gibco/BRL). Phytohemagglutinin-activated
761
PBMC were kept in culture for 2 days prior to addition of
762
Tat protein. Isolation and treatment of PBMC were
763
performed by following the guidelines of the Centers for
764
Disease Control. Approximately 5 × 10 6PBMC were used for
765
treatment of wild type and K41A Tat mutant (100 ng/ml)
766
proteins. After an initial incubation for one hr with Tat
767
proteins, cells were washed and cultured in complete
768
media for 24 hrs, prior to western blots. pCEP4, eTat
769
cells were HeLa cells stably transfected with either a
770
backbone control plasmid (pCEP4; Invitrogen) or a plasmid
771
expressing Tat (1-86) with a C-terminal epitope tag
772
(eTat) [ 103 ] . HeLa cell lines containing either the
773
control or eTat plasmid were selected by single-cell
774
dilution. Both cell types were selected and maintained
775
under 200 μg of hygromycin per ml. Verification of Tat
776
transcriptional activity was achieved by electroporation
777
of reporter plasmids as previously described [ 103 ]
778
.
779
780
781
Cell cycle analysis
782
Hela cells were blocked with hydroxyurea (Hu) (2 mM)
783
for 14 h. Following the block, cells were released by
784
being washed twice with phosphate-buffered saline (PBS)
785
and by the addition of complete medium. All suspension
786
cells were treated with 1% serum for 48 hrs prior to
787
addition of Hu. Supernatants were collected and analyzed
788
by an IL-8 ELISA according to the manufacturer's
789
instructions (Biosource International). For controls,
790
each sample, approximately 1 × 10 6cells was processed
791
for cell sorting. Cells were washed with PBS and fixed by
792
addition of 500 μl of 70% ethanol. For
793
fluorescence-activated cell sorting (FACS) analysis,
794
cells were stained with a cocktail of propidium iodide
795
(PI) buffer (PBS with Ca2+ and Mg2+, RNase A [10 μg/ml],
796
NP-40 [0.1%], and PI [50 μg/ml]) followed by cell-sorting
797
analysis. FACS data acquired were analyzed by ModFit LT
798
software (Verity Software House, Inc.).
799
800
801
Cell extract preparation and immunoblotting
802
All cells were cultured to mid-log phase of growth,
803
washed with PBS without Ca 2+and Mg 2+, and lysed in a
804
buffer containing 50 mM Tris-HCl (pH 7.5), 120 mM NaCl, 5
805
mM EDTA, 50 mM NaF, 0.2 mM Na
806
3 VO
807
4 , 1 mM DTT, 0.5% NP-40 and protease
808
inhibitors (Protease inhibitor cocktail tablets,
809
Boehringer Mannheim, one tablet per 50 ml). The lysate
810
was incubated on ice for 15 min, and microcentrifuged at
811
4°C for 10 min. Total cellular protein was separated on
812
4-20% Tris-glycine gels (Novex, Inc.) and transferred to
813
a polvinylidene difluoride (PVDF) membranes (Immobilon-P
814
transfer membranes; Millipore Corp.) overnight at 0.08 A.
815
Following the transfer, blots were blocked with 5%
816
non-fat dry milk in 50 ml of TNE 50 (100 mM Tris-Cl [pH
817
8.0], 50 mM NaCl, 1 mM EDTA) plus 0.1% NP-40. Membranes
818
were probed with a 1:200-1:1000 dilution of antibodies at
819
4°C overnight, followed by three washes with TNE 50 plus
820
0.1% NP-40. All antibodies used in this study were
821
purchased from Santa Cruz Biotechnology. The next day,
822
blots were incubated with 10 ml of 125I-protein G
823
(Amersham, 50 μl/10 ml solution) in TNE 50 plus 0.1%
824
NP-40 for 2 hrs at 4°C. Finally, blots were washed twice
825
in TNE 50 plus 0.1% NP-40 and placed on a PhosphorImager
826
cassette for further analysis.
827
828
829
Total RNA purification
830
Cells were grown to mid-log phase of growth (5.0 × 10
831
6), pelleted, and washed twice with cold D-PBS without Ca
832
2+/Mg 2+. Total RNA was extracted on ice using Trizol
833
Reagent (Life Technologies, Inc.). Purified RNA was then
834
analyzed on a 1% agarose gel for quality and quantity
835
prior to each experiment.
836
837
838
Glass slide microarray
839
Gene expression analysis was performed using
840
Micromax™: Human cDNA Microarray System I (cat# MPS101,
841
NEN Life Science Products). On a glass microarray slide,
842
2400 know human genes were arrayed into 4 separate grids
843
(A, B, C, D), containing 600 genes each (gene description
844
and location on microarrays available at NEN website:
845
www.nenlifesci.com). All human genes were ~2200 bp cDNAs,
846
and were characterized from 50+ human cDNA libraries
847
(AlphaGene, Inc., Woburn, MA). In addition to the human
848
genes, three plant control genes were spotted on each
849
grid and were utilized to balance the Cyanine-3 (Cy-3)
850
and Cyanine-5 (Cy-5) fluorescence signals.
851
A total of 8 μg each of H9 (control sample) and H9/Tat
852
(test sample) mRNAs were reverse transcribed into Biotin
853
and Dinitrophenyl (DNP) labeled cDNA, respectively. After
854
cDNA quality and quantities were analyzed, both cDNAs
855
were then pooled and simultaneously hybridized overnight
856
at 65°C onto the glass microarray. The next day, the
857
microarray slide was serially washed in 0.5× SSC (Sodium
858
Citrate-Sodium Chloride) + 0.01% SDS (Sodium Dodecyl
859
Sulfate), 0.06× SSC + 0.01% SDS, and 0.06× SSC. Next, the
860
Tyramide Signal Amplification (TSA™) was then used to
861
amplify the Cy-3 and Cy-5 signals using antibody-enzyme
862
conjugates, α-DNP-Horseradish peroxidase (HRP) and
863
α-Streptavidin-HRP with Tyramide linked Cy-3 and Cy-5.
864
Screening and data analysis was performed by NEN.
865
866
867
cDNA filter hybridization
868
Gene expression of CEM and ACH2 were performed using
869
Atlas Human cDNA Expression Array (Clontech Laboratories
870
Inc., Palo Alto, CA) according to the manufacturer's
871
directions. One μg of poly A +RNA each was DNase I
872
treated, purified using a CHROMA SPIN-200 column, and
873
reverse transcribed into 32P-labeled cDNA. The CHROMA
874
SPIN-200 column was used to purify the 32P-labeled cDNA
875
from unincorporated 32P-labeled dNTPs and small (<0.1
876
kb) cDNA fragments. Each sample was then hybridized to a
877
human cDNA expression array overnight with continuous
878
agitation at 68°C. The next day, the array was washed
879
three times with gentle agitation, first wash with 2× SSC
880
+ 1% SDS and the last two washes with 0.1× SSC + 0.5% SDS
881
at 37°C. Array was exposed to a PhosphorImager Cassette
882
and analyzed using ImageQuant software.
883
884
885
Northern blots
886
Total cellular RNA was extracted using the RNAzol
887
reagent (Gibco/BRL). Total RNA (20 μg) was isolated from
888
various cells and ran on a 1% formaldehyde-agarose gel
889
overnight at 75 V, transferred onto a 0.2 μm
890
nitrocellulose membrane (Millipore Inc.), UV
891
cross-linked, and hybridized overnight at 42°C with
892
32P-end-labeled 40 mer oligo probes including p21/Waf1,
893
C-myc, Pro-thymosin, Actin, Tat, and Ubiquitin
894
(Loftstrand, Gaithersburg, Md.). Next day, membranes were
895
washed two times for 15 min each, with 10 ml of 0.2%
896
SDS-2XSSC at 37°C, exposed, and counted on PhosphorImager
897
Cassette.
898
899
900
Viral infection and ELISA assay
901
Both H9 and H9/Tat cells were infected in the presence
902
of 10 ug of polybrene. For PBMC infections, PHA activated
903
PBMCs were kept in culture for 2 days prior to each
904
infection. Isolation and treatment of PBMCs were
905
performed by following guidelines from the CDC
906
(Isolation, culture, and identification of HIV,
907
Procedural Guide, July 1991, Atlanta, GA). Approximately
908
2 × 10 6of H9 or H9/Tat cells, and 5 × 10 6PBMC cells
909
were infected with either an HXB-2 (CXCR4, T-tropic), or
910
BaL (CCR5, Macrophage-tropic) at 5 ng of p24 gag antigen/
911
HIV-1 strain. Both viral isolates were obtained from the
912
NIH AIDS research and reference reagent program. After 8
913
hrs of infection, cells were washed and fresh media was
914
added. Samples were collected every 3 rdday and stored at
915
-20 C for p24 gag ELISA. Media from HIV-1 infected cells
916
were centrifuged to pellet the cells and supernatants
917
were collected, and diluted to 1:100 to 1:1000 in RPMI
918
1640 prior to ELISA. The p24 gag antigen level was
919
analyzed by HIVAG™-1 Monoclonal antibody Kit (Abbott
920
Laboratories, Diagnostics Division).
921
922
923
924
Authors' Contributions
925
CF, and FS carried out the ACH2 and H9/Tat microarrays.
926
LD, CE, IZ, CGL, and KW aided in westerns, northerns, p24
927
and ELISA assays. AM, KK, SB, AP, and FK aided in data
928
interpretation, Bioinformatics, literature searches and
929
writing the manuscript.
930
931
932
933
934