ArticlePDF Available

Evidence for Increased T Cell Turnover and Decreased Thymic Output in HIV Infection

Authors:

Abstract and Figures

The effects of HIV infection upon the thymus and peripheral T cell turnover have been implicated in the pathogenesis of AIDS. In this study, we investigated whether decreased thymic output, increased T cell proliferation, or both can occur in HIV infection. We measured peripheral blood levels of TCR rearrangement excision circles (TREC) and parameters of cell proliferation, including Ki67 expression and ex vivo bromodeoxyuridine incorporation in 22 individuals with early untreated HIV disease and in 15 HIV-infected individuals undergoing temporary interruption of therapy. We found an inverse association between increased T cell proliferation with rapid viral recrudescence and a decrease in TREC levels. However, during early HIV infection, we found that CD45RO-CD27high (naive) CD4+ T cell proliferation did not increase, despite a loss of TREC within naive CD4+ T cells. A possible explanation for this is that decreased thymic output occurs in HIV-infected humans. This suggests that the loss of TREC during HIV infection can arise from a combination of increased T cell proliferation and decreased thymic output, and that both mechanisms can contribute to the perturbations in T cell homeostasis that underlie the pathogenesis of AIDS.
Content may be subject to copyright.
of June 14, 2013.
This information is current as
Decreased Thymic Output in HIV Infection
Evidence for Increased T Cell Turnover and
Richard T. Davey and Richard A. Koup
Michael W. Baseler, Philip Keiser, Douglas D. Richman,
Christian Yoder, Joseph W. Adelsberger, Randy A. Stevens,
Little, Richard Lempicki, Julia A. Metcalf, Joseph Casazza,
Daniel C. Douek, Michael R. Betts, Brenna J. Hill, Susan J.
http://www.jimmunol.org/content/167/11/6663
2001; 167:6663-6668; ;J Immunol
References
http://www.jimmunol.org/content/167/11/6663.full#ref-list-1
, 20 of which you can access for free at: cites 50 articlesThis article
Subscriptions
http://jimmunol.org/subscriptions
is online at: The Journal of ImmunologyInformation about subscribing to
Permissions
http://www.aai.org/ji/copyright.html
Submit copyright permission requests at:
Email Alerts
http://jimmunol.org/cgi/alerts/etoc
Receive free email-alerts when new articles cite this article. Sign up at:
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
Immunologists All rights reserved.
Copyright © 2001 by The American Association of
9650 Rockville Pike, Bethesda, MD 20814-3994.
The American Association of Immunologists, Inc.,
is published twice each month byThe Journal of Immunology
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
Evidence for Increased T Cell Turnover and Decreased
Thymic Output in HIV Infection
1
Daniel C. Douek,
2
*
Michael R. Betts,* Brenna J. Hill,* Susan J. Little,
Richard Lempicki,
Julia A. Metcalf,
Joseph Casazza,* Christian Yoder,
§
Joseph W. Adelsberger,
Randy A. Stevens,
Michael W. Baseler,
Philip Keiser,
#
Douglas D. Richman,
**
Richard T. Davey,
and Richard A. Koup*
The effects of HIV infection upon the thymus and peripheral T cell turnover have been implicated in the pathogenesis of AIDS.
In this study, we investigated whether decreased thymic output, increased T cell proliferation, or both can occur in HIV infection.
We measured peripheral blood levels of TCR rearrangement excision circles (TREC) and parameters of cell proliferation, in-
cluding Ki67 expression and ex vivo bromodeoxyuridine incorporation in 22 individuals with early untreated HIV disease and in
15 HIV-infected individuals undergoing temporary interruption of therapy. We found an inverse association between increased
T cell proliferation with rapid viral recrudescence and a decrease in TREC levels. However, during early HIV infection, we found
that CD45RO
CD27
high
(naive) CD4
T cell proliferation did not increase, despite a loss of TREC within naive CD4
T cells. A
possible explanation for this is that decreased thymic output occurs in HIV-infected humans. This suggests that the loss of TREC
during HIV infection can arise from a combination of increased T cell proliferation and decreased thymic output, and that both
mechanisms can contribute to the perturbations in T cell homeostasis that underlie the pathogenesis of AIDS. The Journal of
Immunology, 2001, 167: 6663–6668.
T
cell depletion of CD4
in HIV infection can arise as a
result of increased destruction and/or reduced production
of T cells through a number of mechanisms, none of
which are mutually exclusive, and for each of which there is ex-
perimental evidence (1–3). T cells can be destroyed by direct or
indirect virus-induced mechanisms, or through Ag-specific CTL-
mediated lysis (4–9). Reduced T cell production can result from
diminished peripheral expansion of pre-existing T cells or inhibi-
tion of de novo generation of naive T cells from thymocytes or
hematopoietic progenitor cells (8, 10–18). Sequestration of cells in
lymphoid tissues may also affect peripheral T cell numbers (19
21). The recovery of T cell numbers during highly active antiret-
roviral therapy (HAART)
3
can occur through a number of different
mechanisms, which may result in the reconstitution of qualitatively
different immune function. Peripheral expansion or redistribution
of pre-existing T cells (17–20, 22) will result in a T cell repertoire
that reflects that already marred by HIV infection, whereas de novo
generation of new naive T cells from the thymus (23–27) will
reconstitute a more diverse T cell repertoire (28, 29).
In vivo bromodeoxyuridine (BrdU) incorporation studies in
SIV-infected monkeys showed increased turnover in all T cell pop-
ulations, with memory T cells affected more than naive (7, 30).
Studies using expression of the Ki67 nuclear Ag as a marker of cell
proliferation indicated that total T cell turnover increased in naive
and memory subsets during infection (31). This suggested that
CD4
T cell loss was due to interference of the virus with “T cell
renewal capacity” rather than with peripheral production, and that
redistribution accounted for increased CD4
T cell numbers dur
-
ing treatment (31). However, a more recent study (32) also using
Ki67 showed that turnover rate, but not proliferation, increased in
CD4
T cells, suggesting their increased death and decreased re
-
newal (32). In vivo labeling with deuterated glucose confirmed
some of these findings, showing that HIV infection caused a de-
crease in memory (but not naive) CD4
and CD8
T cell half-life
with a compensatory increase in production of CD8
but not
CD4
T cells (33).
The measurement of TCR rearrangement excision circles
(TREC) has been used to assess thymic output in individuals with
and without HIV infection (23, 26, 34–37), and after hematopoi-
etic stem cell transplantation (29, 38, 39). In the majority of indi-
viduals with untreated HIV-infection, TREC levels were below
normal, but increased after viral suppression with HAART (23, 26,
37). This was taken to indicate that the thymus, in both adults and
children, is suppressed by HIV infection, but contributes to T cell
*Vaccine Research Center and
Laboratory of Immunoregulation, Clinical and Mo
-
lecular Retrovirology Section, National Institute of Allergy and Infectious Diseases,
Department of Experimental Transplantation and Immunology, Medicine Branch,
National Cancer Institute, and
§
Critical Care Medicine Department, Warren Mag
-
nusen Clinical Center, National Institutes of Health, Bethesda, MD 20892;
Depart
-
ment of Medicine, University of California, San Diego, CA 92103;
Science Appli
-
cations International Corporation-Frederick, Clinical Services Program, Frederick
Cancer Research and Development Center, Frederick, MD 21702;
#
Department of
Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
75390; and **San Diego Veterans Affairs Medical Center, La Jolla, CA 92093
Received for publication June 5, 2001. Accepted for publication September 24, 2001.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by the National Institutes of Health (Grants AI35522 and
AI43638 to R.A.K. and Grant AI43638), by the University of California (San Diego)
Center for AIDS Research (Grants AI36214 and AI291674), by the Research Center
for AIDS and HIV infection of the San Diego Veterans Affairs Healthcare System (to
D.D.R.), by the Leukemia and Lymphoma Society of America (Translational Re-
search Grant 6540-00), and by amFAR (Grant 02680-28-RGV to D.C.D.). This
project has also been funded in part with Federal funds from the National Cancer
Institute under Contract NO1-CO-56000.
2
Address correspondence and reprint requests to Dr. Daniel C. Douek, Vaccine Re
-
search Center, Room 3509, National Institute of Allergy and Infectious Diseases,
National Institutes of Health, 40 Convent Drive, Bethesda, MD 20892; E-mail ad-
dress: ddouek@mail.nih.gov
3
Abbreviations used in this paper: HAART, highly active antiretroviral therapy;
BrdU, bromodeoxyuridine; TREC, TCR rearrangement excision circles.
Copyright © 2001 by The American Association of Immunologists 0022-1767/01/$02.00
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
reconstitution during HAART. In this study, we sought to deter-
mine whether increased T cell turnover, decreased thymic output,
or both occur in HIV infection. We measured peripheral blood
TREC levels and parameters of CD4
and CD8
T cell prolifer
-
ation, including Ki67 expression and ex vivo BrdU incorporation,
in 22 individuals with early untreated HIV disease (312 mo after
infection), and in 15 successfully treated HIV-infected individuals
who underwent temporary interruption of therapy.
Materials and Methods
Human subjects
Twenty-two patients with early HIV infection (312 mo after seroconver-
sion) were seen at the University of Texas Southwestern Medical Center
and had not been on antiretroviral drugs at the time of blood draw. CD4
T cell counts were 220-1080 cells/
l (mean 602) and viral loads were
400 to 7.5 10
4
RNA copies/ml. Fifteen patients were asymptomatic
HIV-infected adults with baseline CD4
T cell counts of 350 cells/
l
who had been on continuous HAART for a minimum period of 1 year, with
viral loads consistently below the limits of detection for at least that period
of time (40). On day 0, patients discontinued all antiretroviral drugs, and
resumed drugs when any of the following three conditions was met: the
CD4
T cell count declined at least 25% from the mean of three baseline
determinations, their viral load increased to 5000 RNA copies/ml, or the
patient resumed drug treatment independently. Viral loads were measured
by Amplicor assay (Roche, Basel, Switzerland). Studies were approved by
the Institutions review boards, and patients gave informed consent.
Measurement of TREC in MACS-sorted cells
Quantication of TREC in sorted CD4
and CD8
T cells was performed
by quantitative PCR with an ABI7700 system (PerkinElmer/Cetus, Nor-
walk, CT) as previously described (29). PBMC were separated into CD4
and CD8
cells using MACS microbeads (Miltenyi Biotec, Auburn, CA).
Cells were lysed in proteinase K (Boehringer Mannheim, Indianapolis, IN)
and PCR was performed on 5
l of cell lysate (50,000 cells). A standard
curve was plotted, and TREC values for samples were calculated by the
ABI7700 software. Samples were analyzed in duplicate. TREC levels are
expressed as TREC per microgram of DNA (1
g of genomic DNA is
equivalent to 150,000 cells). Cell lysates have been checked for consis-
tency of DNA content using
-actin and CCR5 control PCR; interassay
variability was found to be less than 13% of mean for the same sample in
20 different assays (data not shown).
Ex vivo BrdU uptake analysis
Blood samples were incubated with 100
M BrdU for4hat37°C. Cell
surface staining was performed using Abs to CD3, CD45RO, CD4, and/or
CD8 (BD Biosciences, San Jose, CA). Cells were treated with OptiLyse
(Immunotech, Westbrook, ME) for 10 min at room temperature, then with
1% paraformaldehyde and 1% Tween 20 in PBS for 15 min at 37°C.
Cellular DNA was denatured with 100U DNase-I (Boehringer Mannheim)
for 30 min and was then stained with anti-BrdU-FITC (BD Biosciences).
Events (50,000100,000) were collected ow cytometrically, resulting in a
sensitivity of 0.01% BrdU
events, and were analyzed in parallel with
unlabeled cells from the same individual and this value was subtracted
from the value obtained for BrdU-labeled cells. Data are expressed as the
fold change in the percentage of BrdU
cells to avoid large baseline dif
-
ferences in absolute values between individuals. However, similar statis-
tical signicance was obtained when the fold change in absolute BrdU
cells was used (data not shown).
Naive T cell Ki67 analysis and FACS sorting
Analysis was performed by surface staining cells for either CD4/CD45RO/
CD27 or CD8/CD45RO/CD27 (BD Biosciences), followed by xation/
permeabilization and intracellular staining for Ki67 (BD Biosciences).
Cells were analyzed by four-color ow cytometry using a FACSCalibur.
Ki67 expression was measured in both CD45RO
CD27
high
(naive) and
CD45RO
(memory/effector) CD4
and CD8
T cells using Paint-a-Gate
cluster analysis. For FACS sorting of naive and memory T cells, cells were
stained as above (but without Ki67), and were sorted for TREC analysis
using a FACSort (BD Biosciences).
Statistical analyses
Two-tailed Mann-Whitney U test and Spearmans rank correlation coef-
cients were performed using SAS and Prism software (SAS Institute, Cary,
NC). Analysis of covariance was used to assess differences in CD4
and
CD8
T cell TREC between HIV-infected individuals and the healthy
controls, adjusting for age. A p value of 0.05 was considered signicant.
Results
TREC levels measure thymic output
TREC frequency in total and in naive T cells has been shown to
decrease with age, after thymectomy, and in HIV infection (23, 26,
3436, 41). However, a recent mathematical model has suggested
that this decrease in TREC solely reects a theoretical increase in
the naive T cell division rate, and not decreased thymic output (42).
Therefore, we sought to test this experimentally by measuring
changes in T cell division with age using Ki67 expression as a sur-
rogate marker of cell proliferation. Fig. 1 shows that Ki67 expression
does not increase in either CD4
or CD8
CD45RO
CD27
high
(naive) T cells in healthy individuals aged between 23 and 88 years
of age (during which period the most rapid drop in naive T cell
TREC is seen; Refs. 23 and 26). Furthermore, it has recently been
shown that although TREC decrease after thymectomy, there is no
increase in CD27
high
(naive) or CD45RO
memory T cell Ki67
expression (41). The fact that Ki67 may be raised in nonprolifer-
ating or activated cells (3, 43) does not affect this analysis because
the aim in this study was to exclude any increases in proliferation.
Thus, a decrease in TREC levels can indeed reect a decrease in
thymic output.
TREC levels and BrdU uptake after interruption of therapy
It has recently been shown that after total thymectomy, TREC
levels begin to fall after only 3 mo (41). Therefore, in HIV infec-
tion, any decrease in TREC related to decreased thymic output
would not be expected to be observed until at least 3 mo after
seroconversion. Consequently, decreases in TREC before 3 mo of
HIV infection would reect primarily increased T cell
proliferation.
To examine this, we initially studied T cell TREC levels in 15
HIV-infected individuals who had been successfully treated with
HAART, had undetectable viral loads, and then underwent inter-
ruption of therapy (40, 44). As previously reported, recrudescence
of viral replication occurred in all the patients within 28 days of
interruption of therapy. This allowed us to longitudinally assess
changes in TREC and proliferation during a rapid rise in HIV
levelsa situation reminiscent of acute HIV infection. As viral
load rose, both CD4
and CD8
T cell TREC levels decreased
concomitantly. We then performed ex vivo BrdU incorporation to
determine whether this fall in TREC was, in part, secondary to
increased T cell proliferation. The correlation between the S-phase
BrdU fraction and viral load has been previously described (44). In
FIGURE 1. T cell Ki67 expression and age. The percentage of Ki67
CD45RO
CD27
high
(naive) and CD45RO
(memory) CD4
and CD8
T
cells is shown for healthy individuals aged between 23 and 88 years.
6664 THYMIC OUTPUT IN HIV INFECTION
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
the interval between interruption and resumption of therapy, there
was a signicant negative correlation between the change in the
percentage of BrdU
CD4
T cells and the change in CD4
T cell
TREC levels (r ⫽⫺0.6, p 0.02, and 95% condence interval
0.86 to 0.09; Fig. 2). Thus, as CD4
T cell proliferation in-
creased with a concomitant increase in viral load (40), TREC lev-
els decreased. The change in the percentage of BrdU
CD8
T
cells also varied inversely with the change in CD8
T cell TREC
levels. However, this relationship was not statistically signicant
for this sample size (r ⫽⫺0.3, p 0.3, and 95% condence
interval ⫽⫺0.720.31), and a larger study sample would be re-
quired to establish a statistically signicant relationship. It is a
possibility that preferential redistribution of TREC-containing
cells out of the peripheral circulation could cause the decrease in
TREC. Of course, these data do not rule out a concomitant de-
crease in thymic output; it is simply not possible to differentiate the
effects of thymic output and T cell proliferation. However, bearing
in mind the delayed effects of thymectomy on TREC levels (41),
the rapid fall in TREC during an acute rise in HIV load more likely
reects increased T cell proliferation than decreased thymic
output.
TREC levels and Ki67 expression in early HIV infection
Inhibition of thymic function should become detectable by 3 mo
after HIV infection. To differentiate experimentally between thy-
mic inhibition and increased peripheral T cell proliferation, it is
necessary to measure an independent marker of T cell proliferation
in naive and memory T cells, and to measure or calculate the
TREC content of the naive T cell pool. If naive T cell proliferation
is constant, then changes in TREC levels reect changes in the
supply of naive T cells. Therefore, we measured TREC levels and
Ki67 expression in naive and memory CD4
and CD8
T cell
subsets in a separate group of 22 patients with early (312 mo after
seroconversion) untreated HIV infection. Ki67 expression was
used, rather than BrdU incorporation, because these were cryopre-
served samples. It should be stressed, however, that the number of
Ki67
cells and the S-phase fraction (BrdU
cells after a 4-h in
vitro pulse) are not equivalent measures of T cell activation, and
many more cells express Ki67 than are actually in S-phase at any
particular instant in time. The longevity of Ki67 expression after
mitosis remains unclear.
Fig. 3 shows that both CD4
and CD8
T cell TREC were
signicantly lower than in uninfected age-matched controls (p
0.008 and 0.001, respectively). For measurement of Ki67 expres-
sion in naive and memory T cells, subsets were very carefully
dened so that the naive subset would contain few cells outside the
CD27
CD45RO
population. Fig. 4 shows that the percentage of
Ki67
CD4
and Ki67
CD8
CD45RO
(memory) T cells was
signicantly increased in HIV-infected individuals compared with
uninfected individuals (5.7-fold and 6.9-fold, respectively; p
0.0001 for both). We also conrmed that the percentage of
Ki67
CD45RO
(memory) T cells was signicantly higher than
that of naive T cells within the infected and uninfected groups
(CD4, 17.4-fold and p 0.0001; CD8, 5.2-fold and p 0.0003;
CD4, 1.9-fold and p 0.0031; CD8, 2.7-fold and p 0.0017,
infected and uninfected, respectively). However, although the per-
centage of Ki67
CD8
CD45RO
CD27
high
(naive) T cells was
FIGURE 3. Early HIV infection and TREC levels. TREC levels in
sorted CD4
and CD8
T cells from uninfected individuals, (E) and un-
treated individuals with early (312 mo after seroconversion) HIV infec-
tion (F) are shown. Best-t exponential regression curves for uninfected
individuals are shown.
FIGURE 4. T cell Ki67 expression in early HIV infection. The percent-
age of CD4
and CD8
CD45RO
CD27
high
(naive) and CD45RO
(memory) T cells that are Ki67
in uninfected individuals () and indi-
viduals with early HIV infection () are shown. The top, bottom, and line
through the middle of the box correspond to the 75th percentile, 25th per-
centile, and 50th percentile (median), respectively. The whiskers extend
from the 10th percentile to the 90th percentile.
FIGURE 2. Correlation between TREC levels and BrdU incorporation
in treated HIV-infected individuals after therapy interruption. Shown are
the relationships between the fold change in the percentage of
BrdU
CD4
and BrdU
CD8
T cells, and the fold change in CD4
and
CD8
T cell TREC levels in the interval between interruption of therapy
and its resumption with high viral load. Best-t exponential regression
curves are shown.
6665The Journal of Immunology
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
also signicantly increased in HIV-infected individuals (3.6-fold,
p 0.0002), the percentage of Ki67
CD4
CD45RO
CD27
high
(naive) T cells did not differ signicantly from uninfected individ-
uals (p 0.064), and was in fact slightly lower. Thus, even though
Ki67 expression may indicate T cell proliferation and/or activa-
tion, these data suggested that the decrease in CD4
T cell TREC
during HIV infection could not be the result of increased turnover
of naive CD4
T cells.
Naive CD4
T cell activation in early HIV infection
Although we found no increase in naive CD4
T cell Ki67 ex
-
pression in early HIV infection, other studies have shown it to be
increased (31). The phenotypic denition of naive T cells as
CD45RO
CD27
high
by ow cytometry reveals that HIV-infected
subjects contain more T cells with a phenotype between that of
true naive and effector/memory cells than uninfected individuals
(Fig. 5). These have been termed transitional cells (45). There-
fore, we reanalyzed the data shown in Fig. 4, incorporating a small
proportion of transitional CD4
T cells into the naive T cell gate,
and then measuring Ki67 expression. We found that the inclusion
of only 5% more transitional cells apparently increased naive
CD4
T cell Ki67 expression 4.7-fold (range 416) in the HIV-
infected subjects, but only 1.7-fold (range 1.32.2) in the unin-
fected subjects. This difference was statistically signicant (p
0.01), and leads to the misleading conclusion that naive CD4
T
cell Ki67 expression is increased in HIV-infected individuals. An
example of the effect of inclusion of transitional T cells on naive
CD4
T cell Ki67 expression is shown in Fig. 5. These T cells may
have recently been naive cells that are transitioning to activated
cells and that will proliferate. Thus, accurate measurement of ac-
tivation and proliferation in naive CD4
T cells requires rigorous
phenotypic denition of this population by ow cytometry.
Naive T cell TREC levels in early HIV infection
Therefore, to determine whether thymic output was decreased in
early HIV infection in the context of unchanged naive CD4
T cell
proliferation (and also to exclude memory T cell expansions as a
cause of decreased total T cell TREC), we calculated naive T cell
TREC from the total measured TREC and the percentage of naive
T cells determined by ow cytometry. Our calculation assumed
that the contribution of TREC from memory T cells was negligi-
ble. This is a valid assumption, as we have measured TREC in
highly FACS-puried CD4
T cell populations from 12 individ
-
uals and have found that CD45RO
(memory) T cells have, on
average, only 2% of the TREC content of CD45RO
CD27
high
(naive) T cells in the same individual. Furthermore, as an example
of the concordance between calculated and actual measured naive
T cell TREC, we FACS sorted CD45RO
CD27
high
(naive) T cells
in one individual, measured TREC directly, and compared this
result with TREC calculated from unsorted T cells and the naive T
cell percentage, as shown in Tables I and II.
We found that both CD4
and CD8
naive T cell TREC in
early HIV infection were signicantly lower than in uninfected
age-matched controls ( p 0.006 and 0.001, respectively; Fig. 6a).
Because the percentage of Ki67
CD8
CD45RO
CD27
high
(na
-
ive) T cells was increased in HIV-infected individuals, the reduced
naive CD8
TREC levels could have been solely or partly due to
increased naive CD8
T cell proliferation. However, the percent
-
age of Ki67
CD4
CD45RO
CD27
high
(naive) T cells was not
increased, and therefore, the decrease in TREC within
CD45RO
CD27
high
(naive) CD4
T cells could not have been
due to increased proliferation of cells in this compartment. This
suggests that the decreased naive CD4
T cell TREC levels caused
by HIV infection should be reversible when virus replication is
suppressed. To conrm this, we longitudinally followed, after ini-
tiation of HAART, the CD45RO
CD27
high
(naive) CD4
T cell
TREC levels of ve patients who had low TREC before therapy.
After initiation of HAART, which reduced viral loads to undetect-
able levels, CD45RO
CD27
high
(naive) CD4
T cell TREC rose
to normal levels, suggesting that changes in thymic output were
responsible for the changes in TREC within the naive CD4
T
cells before and after HAART (Fig. 6b).
Discussion
There is a considerable body of evidence that HIV can infect the
thymus, both in vitro and in vivo, and compromise its function
(1014, 16, 46, 47). Recent studies of CD4
T cell depletion and
reconstitution in HIV infection have tended to appreciate the mul-
tifactorial nature of immune homeostasisthe composition of na-
ive and memory/effector pools in blood and lymphoid tissue, the
role of the thymus, and the effect of clinical stage of the disease
FIGURE 5. CD45RO
CD27
high
(naive) CD4
T cell Ki67 expression
increases with the inclusion of transitional T cells. Flow cytometric anal-
ysis is shown for two 29-year-old individuals, with the uninfected one on
the left and the HIV-infected one on the right. The panels show the de-
nition of naive T cells by CD27 and CD45RO and then the percentage of
Ki67
cells within that naive population. The top panels are tightly gated
on CD4
CD27
high
CD45RO
naive small lymphocytes, the middle panels
have a wider gate including transitional cells, and the bottom panels show
the result for a wider small lymphocyte gate. The percent Ki67
naive
CD4
T cells is shown in each panel.
Table I. TREC levels per 10
5
cells in CD45RO
CD27
high
(naive) and CD45RO
(memory) T cell subsets measured in sorted T cells in 12
individuals
Subject
TREC per 100,000 CD4
T cells
1 2 3 456789101112
CD45RO
CD27
high
9,106 5,271 24,072 4,224 356 5,960 906 8,256 418 496 1,427 2,209
CD45RO
13 0 30 34 0 168 90 968 0 0 0 0
6666 THYMIC OUTPUT IN HIV INFECTION
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
(32, 33, 42, 46, 48). The use of BrdU incorporation, Ki67 expres-
sion, stable isotope incorporation, and phenotypic denition of T
cell populations has led to a general consensus that CD4
and
CD8
T cell activation and turnover are increased in HIV infec
-
tion, that HIV infection leads to increased death of CD4
T cells,
that there is a defect in the renewal/replacement mechanisms for
CD4
T cells, that these replacement mechanisms have both a
peripheral and thymic component, and that naive CD4
T cell
recovery correlates with thymic size (1, 3, 7, 15, 27, 3033, 42, 44,
45, 49).
The measurement of peripheral blood TREC provides insight
into both thymic function and T cell proliferation. If naive T cell
TREC levels decrease in the absence of an increase in prolifera-
tion, it may be concluded that there is a decrease in the supply of
new TREC
cells, most likely from the thymus. In this study, we
aimed to determine whether decreased thymic output, as well as
increased T cell turnover, occur in HIV infection. In individuals
experiencing a rebound of viral replication after interruption of
therapy, the rapid fall we observed in TREC levels clearly reects
the increase in T cell proliferation. Although not impossible, it is
unlikely that the fall in TREC reects a marked decrease in thymic
output, as it has recently been shown that TREC levels only begin
to decrease 3 mo after total thymectomy in HIV-uninfected indi-
viduals (41).
Therefore, we reasoned that if HIV infection suppressed thymic
output, we would be able to detect this effect at least 3 mo after
infection. Indeed, our results from the analysis of individuals with
early HIV infection 312 mo after seroconversion suggest that
decreased thymic output begins to affect T cell homeostasis by this
stage of the disease. TREC levels were decreased in both CD4
and CD8
T cell subsets, and were also signicantly decreased
when the CD45RO
CD27
high
(naive) T cell TREC were calcu
-
lated. As both CD45RO
(memory) and CD45RO
CD27
high
(naive)
CD8
T cell populations had higher Ki67 expression in HIV-infected
individuals, it was not possible to distinguish between the effects of
increased proliferation and reduced thymic output for CD8
T cells.
However, the percentage of Ki67
CD4
CD45RO
CD27
high
(naive)
T cells did not increase. The nding that TREC within naive CD4
T cells were signicantly lower in infected individuals in the con-
text of unaltered naive CD4
T cell proliferation suggests that the
input of TREC
naive CD4
T cells into the peripheral naive T
cell pool from a source has decreased. The thymus is the most
likely source for such cells (50).
However, some studies have found that the percentage of
Ki67
CD4
naive T cells increased in HIV-infected individuals
(1, 31). The discrepancy between our data and these studies could
be due a number of reasons; for example, our subjects had a higher
mean CD4 counts, and some Ki67
cells might be nondividing (3,
43). However, as we have shown, it is more likely that the incor-
poration of transitional T cells, cells that were naive and have now
become activated, into the phenotypically dened naive T cell sub-
set accounts for the apparent increase in CD4
naive T cell acti
-
vation and/or proliferation in such studies.
Thus, our data show that by 3 mo after HIV infection, a decrease
in TREC within CD45RO
CD27
high
(naive) CD4
T cells is clearly
detectable in the absence of an increase in CD45RO
CD27
high
(na
-
ive) CD4
T cell proliferation, which suggests a decrease in thy
-
mic output. It is possible that the decrease in TREC could be due
to members of the naive pool entering cell cycle, losing TREC due
to dilution, and then reverting to a naive phenotype. However,
there is no evidence in humans for activated or memory CD4
T
cells reverting to a CD45RO
CD27
high
phenotype. The effect of
HIV on thymic function was further conrmed with the observa-
tion that in those individuals with low TREC, CD45RO
CD27
high
(naive) CD4
T cell TREC increased with suppression of virus on
HAART. This suggests that the thymus can recover from its sup-
pression during HIV infection. It is important to note that an in-
crease in naive T cell TREC can only occur in the context of active
thymic output. Therefore, even if decreases in CD4
T cell TREC
occurred due to proliferation and the thymus remained unaffected
by HIV, the increase in naive T cell TREC during HAART indi-
cates that the thymus contributes to immune reconstitution. An
appreciation of the relative roles of the thymus and peripheral T
cell pool in immune reconstitution in HIV infection may provide a
framework for the rational design of interventions that accelerate
and improve the nature of T cell reconstitution in HIV-infected
individuals.
Acknowledgments
We thank Dr. J. Sullivan for samples and Drs. L. Picker and Z. Grossman
for advice.
FIGURE 6. CD45RO
CD27
high
(naive) T cell TREC in early HIV in
-
fection and its treatment. a, CD45RO
CD27
high
(naive) T cell TREC lev
-
els in sorted CD4
and CD8
T cells from uninfected individuals (E) and
individuals with early HIV infection (F), calculated from the percentage of
CD45RO
CD27
high
(naive) T cells present in each population. Best-t
exponential regression curves for uninfected individuals are shown. b, In-
creases during HAART in CD45RO
CD27
high
(naive) CD4
T cell TREC
in ve patients indicated in a who had low TREC before therapy. CD4
T
cell counts per microliter at the start of treatment for the ve patients were:
1, 408; 2, 521; 3, 850; 4, 761; and 5, 416.
Table II. TREC levels per 10
5
cells in CD45RO
CD27
high
(naive) and CD45RO
(memory) T cell subsets
measured and back-calculated in one individual
% CD45RO
CD27
high
Naive CD4
T Cells
MACS-Sorted
CD4
T Cell
TREC
Calculated Naive
CD4
T Cell
TREC
FACS-Sorted Naive
CD4
T Cell TREC
FACS-Sorted Memory
CD4
T Cell TREC
34.06% 692 2,032 2,054 6
6667The Journal of Immunology
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
References
1. Sachsenberg, N., A. S. Perelson, S. Yerly, G. A. Schockmel, D. Leduc,
B. Hirschel, and L. Perrin. 1998. Turnover of CD4
and CD8
T lymphocytes
in HIV-1 infection as measured by Ki-67 antigen. J. Exp. Med. 187:1295.
2. Rowland-Jones, S. 1999. HIV infection: where have all the T cells gone? Lancet
354:5.
3. Grossman, Z., R. B. Herberman, and D. S. Dimitrov. 1999. T cell turnover in SIV
infection. Science 284:555.
4. Ho, D. D., A. U. Neumann, A. S. Perelson, W. Chen, J. M. Leonard, and
M. Markowitz. 1995. Rapid turnover of plasma virions and CD4 lymphocytes in
HIV-1 infection. Nature 373:123.
5. Wei, X., S. Ghosh, M. Taylor, V. Johnson, E. Emini, P. Deutsch, J. Lifson,
S. Bonhoeffer, M. Nowak, B. Hahn, et al. 1995. Viral dynamics in human im-
munodeciency virus type 1 infection. Nature 373:117.
6. Zhang, Z.-Q., D. Notermans, G. Sedgewick, W. Cavert, S. Sietgrefe,
M. Zupancic, S. Gebhard, K. Henry, L. Boies, Z. Chen, M. Jenkins, R. Mills, et
al. 1998. Kinetics of CD4
T cell repopulation of lymphoid tissues after treat
-
ment of HIV-1 infection. Proc. Natl. Acad. Sci. USA 95:1154.
7. Mohri, H., S. Bonhoeffer, S. Monard, A. S. Perelson, and D. D. Ho. 1998. Rapid
turnover of T lymphocytes in SIV-infected rhesus macaques. Science 279:1223.
8. Tenner-Racz, K., H. J. Stellbrink, J. van Lunzen, C. Schneider, J. P. Jacobs,
B. Raschdorff, G. Grosschupff, R. M. Steinman, and P. Racz. 1998. The unen-
larged lymph nodes of HIV-1-infected, asymptomatic patients with high CD4 T
cell counts are sites for virus replication and CD4 T cell proliferation: the impact
of highly active antiretroviral therapy. J. Exp. Med. 187:949.
9. Gandhi, R. T., B. K. Chen, S. E. Straus, J. K. Dale, M. J. Lenardo, and
D. Baltimore. 1998. HIV-1 directly kills CD4
T cells by a Fas-independent
mechanism. J. Exp. Med. 187:1113.
10. Schnittman, S. M., S. M. Denning, J. J. Greenhouse, J. S. Justement, M. Baseler,
B. F. Haynes, and A. S. Fauci. 1990. Evidence for susceptibility of intrathymic
T cell precursors to human immunodeciency virus infection: a mechanism for
T4 (CD4) lymphocyte depletion. Trans Assoc Am Physicians 103:96.
11. Bonyhadi, M., L. Rabin, S. Salimi, D. Brown, J. Kosek, J. McCune, and
H. Kaneshima. 1993. HIV induces thymus depletion in vivo. Nature 363:728.
12. Aldrovandi, G. M., G. Feuer, L. Gao, B. Jamieson, M. Kristeva, I. S. Chen, and
J. A. Zack. 1993. The SCID-hu mouse as a model for HIV-1 infection. Nature
363:732.
13. Jamieson, B., C. Uittenbogaart, I. Schmid, and J. Zack. 1997. High viral burden and
rapid CD4
cell depletion in human immunodeciency virus type 1-infected
SCID-hu mice suggest direct viral killing of thymocytes in vivo. J. Virol. 71:8245.
14. Su, L., H. Kaneshima, M. Bonyhadi, S. Salimi, D. Kraft, L. Rabin, and
J. McCune. 1995. HIV-1-induced thymocyte depletion is associated with indirect
cytopathicity and infection of progenitor cells in vivo. Immunity 2:25.
15. Roederer, M., S. C. De Rosa, N. Watanabe, and L. A. Herzenberg. 1997. Dy-
namics of ne T-cell subsets during HIV disease and after thymic ablation by
mediastinal irradiation. Semin. Immunol. 9:389.
16. Wykrzykowska, J., M. Rosenzweig, R. Veazey, M. Simon, K. Halvorsen,
R. Desrosiers, R. Johnson, and A. Lackner. 1998. Early regeneration of thymic
progenitors in rhesus macaques infected with simian immunodeciency virus.
J. Exp. Med. 187:1767.
17. Fleury, S., R. de Boer, G. Rizzardi, K. Wolthers, S. Otto, C. Welbon, C. Graziosi,
C. Knabenhans, H. Soudeyns, P. Bart, et al. 1998. Limited CD4
T cell renewal in
early HIV-1 infection: effect of highly active antiretroviral therapy. Nat. Med. 4:794.
18. Hellerstein, M., M. B. Hanley, D. Cesar, S. Siler, C. Papageorgopoulos,
E. Wieder, D. Schmidt, R. Hoh, R. Neese, D. Macallan, et al. 1999. Directly
measured kinetics of circulating T lymphocytes in normal and HIV-1-infected
humans. Nat. Med. 5:83.
19. Pakker, N., D. Notermans, R. De Boer, M. Roos, F. De Wolf, A. Hill, J. Leonard,
S. Danner, F. Miedema, and P. Schellekens. 1998. Biphasic kinetics of peripheral
blood T cells after triple combination therapy in HIV-1 infection: a composite of
redistribution and proliferation. Nat. Med. 4:208.
20. Bucy, R. P., R. D. Hockett, C. A. Derdeyn, M. S. Saag, K. Squires, M. Sillers,
R. T. Mitsuyasu, and J. M. Kilby. 1999. Initial increase in blood CD4
lympho
-
cytes after HIV antiretroviral therapy reects redistribution from lymphoid tis-
sues. J. Clin. Invest. 103:1391.
21. Wang, L., J. J. Chen, B. B. Gelman, R. Konig, and M. W. Cloyd. 1999. A novel
mechanism of CD4 lymphocyte depletion involves effects of HIV on resting
lymphocytes: induction of lymph node homing and apoptosis upon secondary
signaling through homing receptors. J. Immunol. 162:268.
22. Walker, R. E., C. S. Carter, L. Muul, V. Natarajan, B. R. Herpin, S. F. Leitman,
H. G. Klein, C. A. Mullen, J. A. Metcalf, M. Baseler, et al. 1998. Peripheral
expansion of pre-existing mature T cells is an important means of CD4
T-cell
regeneration HIV-infected adults. Nat. Med. 4:852.
23. Douek, D. C., R. D. McFarland, P. H. Keiser, E. A. Gage, J. M. Massey,
B. F. Haynes, M. A. Polis, A. T. Haase, M. B. Feinberg, J. L. Sullivan, et al. 1998.
Changes in thymic function with age and during the treatment of HIV infection.
Nature 396:690.
24. McCune, J., R. Loftus, D. Schmidt, P. Carroll, D. Webster, L. Swor-Yim,
I. Francis, B. Gross, and R. Grant. 1998. High prevalence of thymic tissue in
adults with human immunodeciency virus-1 infection. J. Clin. Invest. 101:2301.
25. Poulin, J. F., and R. P. Sekaly. 1999. Function of the thymus in HIV-infected
adults. J. Am. Med. Assoc. 282:219.
26. Zhang, L. Q., S. R. Lewin, M. Markowitz, H.-H. Lin, E. Skulsky, R. Karanicolas,
Y. He, X. Jin, S. Tuttleton, M. Vesanen, et al. 1999. Measuring recent thymic
emigrants in blood of normal persons and HIV-1-infected individuals before and
after effective therapy. J. Exp. Med. 190:725.
27. Smith, K. Y., H. Valdez, A. Landay, J. Spritzler, H. A. Kessler, E. Connick,
D. Kuritzkes, B. Gross, I. Francis, J. M. McCune, and M. M. Lederman. 2000.
Thymic size and lymphocyte restoration in patients with human immunode-
ciency virus infection after 48 weeks of zidovudine, lamivudine, and ritonavir
therapy. J. Infect. Dis. 181:141.
28. Heitger, A., N. Neu, H. Kern, E. R. Panzer-Grumayer, H. Greinix, D. Nachbaur,
D. Niederwieser, and F. M. Fink. 1997. Essential role of the thymus to recon-
stitute naive (CD45RA
) T-helper cells after human allogeneic bone marrow
transplantation. Blood 90:850.
29. Douek, D., R. Vescio, M. Betts, J. Brenchley, B. Hill, L. Zhang, J. Berenson, R.
Collins, and R. Koup. 2000. Assessment of thymic output in adults after haema-
topoietic stem cell transplant and prediction of T cell reconstitution. Lancet 355:
1875.
30. Rosenzweig, M., M. A. DeMaria, D. M. Harper, S. Friedrich, R. K. Jain, and
R. P. Johnson. 1998. Increased rates of CD4
and CD8
T lymphocyte turnover
in simian immunodeciency virus-infected macaques. Proc. Natl. Acad. Sci. USA
95:6388.
31. Hazenberg, M. D., J. W. Stuart, S. A. Otto, J. C. Borleffs, C. A. Boucher,
R. J. de Boer, F. Miedema, and D. Hamann. 2000. T-cell division in human
immunodeciency virus (HIV)-1 infection is mainly due to immune activation: a
longitudinal analysis in patients before and during highly active antiretroviral
therapy (HAART). Blood 95:249.
32. Fleury, S., G. P. Rizzardi, A. Chapuis, G. Tambussi, C. Knabenhans, E. Simeoni,
J. Meuwly, J. Corpataux, A. Lazzarin, F. Miedema, and G. Pantaleo. 2000. Long-
term kinetics of T cell production in HIV-infected subjects treated with highly
active antiretroviral therapy. Proc. Natl. Acad. Sci. USA 97:5393.
33. McCune, J. M., M. B. Hanley, D. Cesar, R. Halvorsen, R. Hoh, D. Schmidt,
E. Wieder, S. Deeks, S. Siler, R. Neese, and M. Hellerstein. 2000. Factors in-
uencing T-cell turnover in HIV-1-seropositive patients. J. Clin. Invest. 105:R1.
34. Jamieson, B. D., D. C. Douek, S. Killian, L. E. Hultin, D. D. Scripture-Adams,
J. V. Giorgi, D. Marelli, R. A. Koup, and J. A. Zack. 1999. Generation of func-
tional thymocytes in the human adult. Immunity 10:569.
35. Poulin, J. F., M. N. Viswanathan, J. M. Harris, K. V. Komanduri, E. Wieder,
N. Ringuette, M. Jenkins, J. M. McCune, and R. P. Sekaly. 1999. Direct evidence
for thymic function in adult humans. J. Exp. Med. 190:479.
36. Hatzakis, A., G. Touloumi, R. Karanicolas, A. Karafoulidou, T. Mandalaki,
C. Anastassopoulou, L. Zhang, J. J. Goedert, D. D. Ho, and L. G. Kostrikis. 2000.
Effect of recent thymic emigrants on progression of HIV-1 disease. Lancet 355:599.
37. Douek, D. C., R. A. Koup, R. D. McFarland, J. L. Sullivan, and K. Luzuriaga.
2000. Effect of HIV on thymic function before and after antiretroviral therapy in
children. J. Infect. Dis. 181:1479.
38. Patel, D. D., M. E. Gooding, R. E. Parrott, K. M. Curtis, B. F. Haynes, and
R. H. Buckley. 2000. Thymic function after hematopoietic stem-cell transplan-
tation for the treatment of severe combined immunodeciency. N. Engl. J. Med.
342:1325.
39. Weinberg, K., B. R. Blazar, J. E. Wagner, E. Agura, B. J. Hill,
M. Smogorzewska, R. A. Koup, M. R. Betts, R. H. Collins, and D. C. Douek.
2001. Factors affecting thymic function after allogeneic hematopoietic stem cell
transplantation. Blood 97:1458.
40. Davey, R. T., Jr., N. Bhat, C. Yoder, T. W. Chun, J. A. Metcalf, R. Dewar,
V. Natarajan, R. A. Lempicki, J. W. Adelsberger, K. D. Miller, et al. 1999. HIV-1
and T cell dynamics after interruption of highly active antiretroviral therapy
(HAART) in patients with a history of sustained viral suppression. Proc. Natl.
Acad. Sci. USA 96:15109.
41. Sempowski, G., J. Thomasch, M. Gooding, L. Hale, L. Edwards, E. Ciafaloni,
D. Sanders, J. Massey, D. Douek, R. Koup, and B. Haynes. 2001. Effect of
thymectomy on human peripheral blood T cell pools in myasthenia gravis. J. Im-
munol. 166:2808.
42. Hazenberg, M., S. Otto, J. Cohen Stuart, M. Verschuren, J. Borleffs, C. Boucher,
R. Coutinho, J. Lange, T. Rinke de Wit, A. Tsegaye, et al. 2000. Increased cell
division but not thymic dysfunction rapidly affects the T-cell receptor excision circles
content of the naive T cell population in HIV-1 infection. Nat. Med. 6:1036.
43. van Oijen, M. G., R. H. Medema, P. J. Slootweg, and G. Rijksen. 1998. Positivity
of the proliferation marker Ki-67 in noncycling cells. Am. J. Clin. Pathol. 110:24.
44. Lempicki, R. A., J. A. Kovacs, M. W. Baseler, J. W. Adelsberger, R. L. Dewar,
V. Natarajan, M. C. Bosche, J. A. Metcalf, R. A. Stevens, L. A. Lambert, et al.
2000. Impact of HIV-1 infection and highly active antiretroviral therapy on the
kinetics of CD4
and CD8
T cell turnover in HIV-infected patients. Proc. Natl.
Acad. Sci. USA 97:13778.
45. Grossman, Z., and W. Paul. 2000. The impact of HIV on naı¨ve T-cell homeosta-
sis. Nat. Med. 6:976.
46. Haase, A. T. 1999. Population biology of HIV-1 infection: viral and CD4
T cell
demographics and dynamics in lymphatic tissues. Annu. Rev. Immunol. 17:625.
47. Haynes, B. F., L. P. Hale, K. J. Weinhold, D. D. Patel, H. X. Liao, P. B. Bressler,
D. M. Jones, J. F. Demarest, K. Gebhard-Mitchell, et al. 1999. Analysis of the
adult thymus in reconstitution of T lymphocytes in HIV-1 infection. J. Clin.
Invest. 103:453.
48. Haynes, B. F., M. L. Markert, G. D. Sempowski, D. D. Patel, and L. P. Hale.
2000. The role of the thymus in immune reconstitution in aging, bone marrow
transplantation, and HIV-1 infection. Annu. Rev. Immunol. 18:529.
49. Grossman, Z., and R. Herberman. 1997. T-cell homeostasis in HIV infection is
neither failing nor blind: modied cell counts reect an adaptive response of the
host. Nat. Med. 3:486.
50. Markert, M. L., A. Boeck, L. P. Hale, A. L. Kloster, T. M. McLaughlin,
M. N. Batchvarova, D. C. Douek, R. A. Koup, D. D. Kostyu, F. E. Ward, et al.
1999. Transplantation of thymus tissue in complete DiGeorge syndrome. N. Engl.
J. Med. 341:1180.
6668 THYMIC OUTPUT IN HIV INFECTION
at NIH Library, National Institutes of Health on June 14, 2013http://www.jimmunol.org/Downloaded from
... These increased levels of T-cell proliferation go hand in hand with significantly decreased T-cell TREC contents in untreated HIV infection -a natural consequence of the fact that TRECs are not copied during cell division (BOX 4, How to measure thymic output?). The fraction of proliferating CD4 + T cells in individuals with untreated HIV has been shown to inversely correlate with CD4 + T-cell numbers, suggesting that T cells proliferate faster to return to normal T-cell numbers (47,78,79). Yet, there is a large body of evidence suggesting that T-cell dynamics during HIV infection are to a large extent driven by the virus itself, and not necessarily by a homeostatic response to low T-cell numbers. ...
... Based on the observation that the average TREC content of CD4 + T cells increases during ART, concomitant with the recovery of naive CD4 + T-cell numbers (75,76) (Figure 3), it has been proposed that, once viral replication is suppressed, increased thymic output plays a key role in the reconstitution of the T-cell pool (79). Computed tomography (CT) scans have shown that the adult thymus can indeed expand after the start of antiretroviral therapy (82)(83)(84). ...
Article
Full-text available
A timely recovery of T-cell numbers following haematopoietic stem-cell transplantation (HSCT) is essential for preventing complications, such as increased risk of infection and disease relapse. In analogy to the occurrence of lymphopenia-induced proliferation in mice, T-cell dynamics in humans are thought to be homeostatically regulated in a cell density-dependent manner. The idea is that T cells divide faster and/or live longer when T-cell numbers are low, thereby helping the reconstitution of the T-cell pool. T-cell reconstitution after HSCT is, however, known to occur notoriously slowly. In fact, the evidence for the existence of homeostatic mechanisms in humans is quite ambiguous, since lymphopenia is often associated with infectious complications and immune activation, which confound the study of homeostatic regulation. This calls into question whether homeostatic mechanisms aid the reconstitution of the T-cell pool during lymphopenia in humans. Here we review the changes in T-cell dynamics in different situations of T-cell deficiency in humans, including the early development of the immune system after birth, healthy ageing, HIV infection, thymectomy and hematopoietic stem cell transplantation (HSCT). We discuss to what extent these changes in T-cell dynamics are a side-effect of increased immune activation during lymphopenia, and to what extent they truly reflect homeostatic mechanisms.
... The reason for better outcomes in children remain unknown, but may be related to differences in the homeostasis of their T cell repertoire. In adults, the maintenance of T cells in the periphery occurs predominantly by redistribution of cells from tissues to blood, with a small and very slow recovery in the naive repertoire, largely due to proliferation of existing T cell clones with limited thymic production (6). In contrast, young children have a very high thymic output of naïve T cells, peaking at 1-2 years, then gradually declining to much lower levels by early adulthood, resulting in dramatic reduction of T cell receptor (TCR) diversity with age (7)(8)(9)(10)(11). ...
Article
It is intriguing that, unlike adults with HIV-1, children with HIV-1 reach a greater CD4+ T cell recovery following planned treatment cessation. The reasons for the better outcomes in children remain unknown but may be related to increased thymic output and diversity of T cell receptor repertoires. HIV-1 infected children from the PENTA 11 trial tolerated planned treatment interruption without adverse long-term clinical, virological, or immunological consequences, once antiretroviral therapy was re-introduced. This contrasts to treatment interruption trials of HIV-1 infected adults, who had rapid changes in T cells and slow recovery when antiretroviral therapy was restarted. How children can develop such effective immune responses to planned treatment interruption may be critical for future studies. PENTA 11 was a randomized, phase II trial of planned treatment interruptions in HIV-1-infected children (ISRCTN 36694210). In this sub-study, eight patients in long-term follow-up were chosen with CD4+ count>500/ml, viral load <50c/ml at baseline: four patients on treatment interruption and four on continuous treatment. Together with measurements of thymic output, we used high-throughput next generation sequencing and bioinformatics to systematically organize memory CD8+ and naïve CD4+ T cell receptors according to diversity, clonal expansions, sequence sharing, antigen specificity, and T cell receptor similarities following treatment interruption compared to continuous treatment. We observed an increase in thymic output following treatment interruption compared to continuous treatment. This was accompanied by an increase in T cell receptor clonal expansions, increased T cell receptor sharing, and higher sequence similarities between patients, suggesting a more focused T cell receptor repertoire. The low numbers of patients included is a limitation and the data should be interpreted with caution. Nonetheless, the high levels of thymic output and the high diversity of the T cell receptor repertoire in children may be sufficient to reconstitute the T cell immune repertoire and reverse the impact of interruption of antiretroviral therapy. Importantly, the effective T cell receptor repertoires following treatment interruption may inform novel therapeutic strategies in children infected with HIV-1.
... Infection recovery is characterized by the clearance of infection and the development of immunological memory facilitated by memory B and T cells, which should prevent the onset of recurrent disease, through rapid cellular proliferation and antibody production upon secondary exposure to the pathogen. 59,60 Infectioninduced thymic damage and DP thymocyte depletion results in fewer SP CD4 and CD8 naïve T cells migrating into the peripheral circulation of HIV infected mice 61 and immature thymocytes escaping the thymus in T. cruzi 62 and Plasmodium berghei infection. 63 These phenomena may also be occurring during CDI. ...
Article
Full-text available
Clostridioides difficile is the most common cause of infectious antibiotic-associated diarrhea, with disease mediated by two major toxins TcdA and TcdB. In severe cases, systemic disease complications may arise, resulting in fatal disease. Systemic disease in animal models has been described, with thymic damage an observable consequence of severe disease in mice. Using a mouse model of C. difficile infection, we examined this disease phenotype, focussing on the thymus and serum markers of systemic disease. The efficacy of bezlotoxumab, a monoclonal TcdB therapeutic, to prevent toxin mediated systemic disease complications was also examined. C. difficile infection causes toxin-dependent thymic damage and CD4⁺CD8⁺ thymocyte depletion in mice. These systemic complications coincide with changes in biochemical markers of liver and kidney function, including increased serum urea and creatinine, and hypoglycemia. Administration of bezlotoxumab during C. difficile infection prevents systemic disease and thymic atrophy, without blocking gut damage, suggesting the leakage of gut contents into circulation may influence systemic disease. As the thymus has such a crucial role in T cell production and immune system development, these findings may have important implications in relapse of C. difficile disease and impaired immunity during C. difficile infection. The prevention of thymic atrophy and reduced systemic response following bezlotoxumab treatment, without altering colonic damage, highlights the importance of systemic disease in C. difficile infection, and provides new insights into the mechanism of action for this therapeutic. Abbreviations: Acute kidney injury (AKI); Alanine Transaminase (ALT); Aspartate Aminotransferase (AST); C. difficile infection (CDI); chronic kidney disease (CKD); combined repetitive oligo-peptides (CROPS); cardiovascular disease (CVD); Double positive (DP); hematoxylin and eosin (H&E); immunohistochemical (IHC); multiple organ dysfunction syndrome (MODS); phosphate buffered saline (PBS); standard error of the mean (SEM); surface layer proteins (SLP); Single positive (SP); wild-type (WT).
... We analyzed TRECs to evaluate their "proliferative history" and state of naïvety. During TCR rearrangement, TRECs are generated, and their prevalence in T cells has been used as a marker of their naïve status and lack of TCR-driven proliferation (35,(42)(43)(44). TREC concentrations were similar between purified naïve CD8 þ T cells from patients with or without an elevated LAG3 IC and purified naïve HD CD8 þ T cells (Fig. 4D). ...
Article
Many cancer patients do not develop a durable response to the current standard of care immunotherapies, despite substantial advances in targeting immune inhibitory receptors. A potential compounding issue, which may serve as an unappreciated, dominant resistance mechanism, is an inherent systemic immune dysfunction that is often associated with advanced cancer. Minimal response to inhibitory receptor (IR) blockade therapy and increased disease burden have been associated with peripheral CD8+ T-cell dysfunction, characterized by suboptimal T-cell proliferation and chronic expression of IRs (eg. Programmed Death 1 [PD1] and Lymphocyte Activation Gene 3 [LAG3]). Here, we demonstrated that approximately a third of cancer patients analyzed in this study have peripheral CD8+ T cells that expressed robust intracellular LAG3 (LAG3IC), but not surface LAG3 (LAG3SUR) due to A Disintegrin and Metalloproteinase domain-containing protein 10 (ADAM10) cleavage. This associated with poor disease prognosis and decreased CD8+ T-cell function, which could be partially reversed by anti-LAG3. Systemic immune dysfunction was restricted to CD8+ T cells, including, in some cases, a high percentage of peripheral naïve CD8+ T cells, and was driven by the cytokine IL6 via STAT3. These data suggest that additional studies are warrented to determine if the combination of increased LAG3IC in peripheral CD8+ T cells and elevated systemic IL6 can serve as predictive biomarkers and identify which cancer patients may benefit from LAG3 blockade.
... For simplicity, we set C 0 equal to one in the following and we assume constant rates d and q. Importantly, recruitment of new clones and total expansion of already existing clones maintain a constant ratio throughout development under these assumptions in line with findings that the fraction of cells with TCR excision circles, which are diluted during peripheral division, is constant during fetal development (Schö nland et al., 2003) and infancy (Douek et al., 2001;Bains et al., 2009). We simulated the model starting from an empty repertoire and found that large clones displayed power-law scaling ( Figure 2B blue lines). ...
Article
The adaptive immune system responds to pathogens by selecting clones of cells with specific receptors. While clonal selection in response to particular antigens has been studied in detail, it is unknown how a lifetime of exposures to many antigens collectively shape the immune repertoire. Here, using mathematical modeling and statistical analyses of T cell receptor sequencing data, we develop a quantitative theory of human T cell dynamics compatible with the statistical laws of repertoire organization. We find that clonal expansions during a perinatal time window leave a long-lasting imprint on the human T cell repertoire, which is only slowly reshaped by fluctuating clonal selection during adult life. Our work provides a mechanism for how early clonal dynamics imprint the hierarchy of T cell clone sizes with implications for pathogen defense and autoimmunity.
... 58 In addition, poor recovery of CD4 + T cells is associated with thymic immigrants and reduced thymic output. 59,60 On the other hand, poor immune reconstitution is associated with reduced bone marrow hematopoiesis. Bone marrow function is abnormal in HIV-infected patients; it is mostly restored after highly active ART, 61,62 but remains abnormal in many patients with poor immune reconstitution. ...
Article
Human immunodeficiency virus type 1 (HIV‐1) infection causes considerable morbidity and mortality worldwide. Although antiretroviral therapy (ART) has largely transformed HIV infection from a fatal disease to a chronic condition, approximately 10%~40% of HIV‐infected individuals who receive effective ART and sustain long‐term viral suppression still cannot achieve optimal immune reconstitution. These patients are called immunological non‐responders, a state associated with poor clinical prognosis. Mucosal‐associated invariant T (MAIT) cells are an evolutionarily conserved unconventional T cell subset defined by expression of semi‐invariant αβ T cell receptor (TCR), which recognizes metabolites derived from the riboflavin biosynthetic pathway presented on major histocompatibility complex (MHC)‐related protein‐1 (MR1). MAIT cells, which are considered to act as a bridge between innate and adaptive immunity, produce a wide range of cytokines and cytotoxic molecules upon activation through TCR‐dependent and TCR‐independent mechanisms, which is of major importance in defense against a variety of pathogens. In addition, MAIT cells are involved in autoimmune and immune‐mediated diseases. The number of MAIT cells is dramatically and irreversibly decreased in the early stage of HIV infection and is not fully restored even after long‐term suppressive ART. In light of the important role of MAIT cells in mucosal immunity and because microbial translocation is inversely associated with CD4+ T cell counts, we propose that MAIT cells participate in the maintenance of intestinal barrier integrity and microbial homeostasis, thus further affecting immune reconstitution in HIV‐infected individuals. This article is protected by copyright. All rights reserved.
Article
Research data presented in the article are based, mainly, on the concept that thymus, together with bone marrow represent the central organ of immune system being the source of all T cell populations that, following their migration from thymus to periphery, participate in development of immune response to any antigens of viral, bacterial and tissue origin, and to any allergens. This difference is principal, as opposed to the bone marrow which produces other members of immune response, i.e., dendritic cells, macrophages, B cells. E.g., the bone marrow also generates the cells which migrate to thymus where they undergo differentiation to the T cells. Over last 50 years, a plethora of data was accumulated on the leading role of immune system in pathogenesis of virtually all socially significant human diseases affecting the modern mankind, including infectious and malignant disorders, atherosclerosis, autoimmune and allergic diseases. Moreover, current studies show that the aberrant functions of different T cell populations play the leading role in pathogenesis of these diseases. These T cell disturbances in peripheral areas of different organs are proven to develop, mainly in the thymic area. Hence, thymus is a producing organ of T cells with altered functional activities which promote pathogenetic changes in these disorders. Currently, the entire set of immunotherapeutic approaches is aimed for correction of disturbances among the same T cells subpopulations at periphery, without taking into account thymic mechanisms which have induced these disturbances before their emigration from thymus. One should, therefore, develop novel methods and approaches to correct these alterations within thymic area.
Article
Full-text available
Background: Lymphopenia, particularly restricted to the T-cell compartment, has been described as one of the major clinical hallmarks in COVID-19 patients and proposed as a disease severity indicator. Although several mechanisms fostering COVID-19-related lymphopenia have been described, including cell apoptosis and tissue homing, the underlying causes of the decline in T-cell count and function are still not completely understood. Objective: Given that viral infections can directly target thymic microenvironment and impair the process of T-cell generation, we sought to investigate the impact of SARS-CoV-2 on thymic function. Methods: We performed molecular quantification of T-cell receptor excision circles (TRECs) and kappa-deleting recombination excision circles (KRECs) to assess, respectively, T- and B-cell neogenesis in SARS-CoV-2-infected patients. We developed in vitro culture system of primary human thymic epithelial cells (TECs) to mechanistically investigate the impact of SARS-CoV-2 on TEC function. Results: We showed that COVID-19 patients had reduced thymic function, which was inversely associated with the severity of the disease. We found that angiotensin-converting enzyme 2 (ACE2), through which SARS-CoV-2 enters the host cells, was expressed by thymic epithelium, and in particular by medullary TECs. We also demonstrated that SARS-CoV-2 can target TECs and downregulate critical genes and pathways associated with epithelial cell adhesion and survival. Conclusions: Our data demonstrate that the human thymus is a target of SARS-CoV-2 and thymic function is altered upon infection. These findings expand our current knowledge of the effects of SARS-CoV-2 infection on T-cell homeostasis and suggest that monitoring thymic activity may represent a useful marker to predict disease severity and progression.
Article
Full-text available
РАС страдает каждый 59-й ребенок. У пациентов с РАС чаще, чем в популяции в целом, присутствуют сопутствующие расстройства, которые могут усугубить течение основного заболевания или повлиять на постановку диагноза. Цель работы – выделить клинико-иммунологические фенотипы течения РАС. Материалом исследования служили образцы крови детей двух групп исследования: дети с РАС (n=100) и клинически здоровые (n=30). На основании наличия сопутствующих заболеваний дети были разделены на 3 фенотипа: судорожный, инфекционный, дермато-респираторный и желудочно-кишечный. В сыворотке крови определяли концентрации цитокинов IL-4, IL-6, IL-10, IFN-γ, IL-17А. Модифицированным методом ИФА с применением методологии Immunohealth™ определяли концентрацию spIgG к 111 пищевым антигенам (пАГ). Оценку показателей когнитивных и психофизиологических показателей у детей проводили с помощью анкеты АТЕС. В результате исследования выделены клинико-иммунологические фенотипы течения РАС, связанные с особым типом пищевой реактивности, цитокиновым профилем, клинической тяжестью психо-физиологических расстройств и сопутствующими коморбидными заболеваниями. Во всех четырех фенотипах установлена повышенная активность синтеза специфических антител, связанного с изученным пАГ гуморального иммунитета, повышенная концентрация суммарного spIgG к пАГ и концентрации spIgG к бобовым продуктам и казеину, значения С-реактивного белка. При этом в судорожном фенотипе (сопутствующие эпилепсия и судороги) максимальные концентрации spIgG установлены к паслёновым продуктам, повышена концентрация IL-10 и снижена концентрация IL-4, снижено содержание сывороточного железа и ферритина. В инфекционном фенотипе (часто болеющие дети) – к зерновым и бродильным продуктам, повышены концентрации IL-10 и IFN-γ и снижена концентрация IL-4, повышены абсолютное и относительное количество лимфоцитов и фибриноген. В дермато-респираторном фенотипе (кожные высыпания) – к молочным продуктам, повышены концентрации IL-4 и IL-17А. В желудочно-кишечном фенотипе зарегистрировано самое высокое количество реакций повышенных IgG к наибольшему спектру пАГ на фоне тенденции изменения в цитокиновом профиле в сторону повышения IFN-γ в соотношениях IFN=γ/IL-4 и IFN=γ/IL-10. Таким образом, выделенные фенотипы течения РАС связаны с влиянием пАГ и отражают особый вариант иммунологического воспалительного патогенеза, что позволяет персонифицировать элиминационные диеты, предложить мероприятия по коррекции и индивидуальной профилактике и, вероятно, рассчитать прогноз течения заболевания.
Article
Full-text available
In their report of 20 February 1998 ([1][1]), H. Mohri et al . analyze the proliferation and death of T cells in normal macaques and in those infected with simian immunodeficiency virus (SIV). With the use of bromodeoxyuridine (BrdU) to label proliferating cells, they observed that the uptake of
Article
Full-text available
The role of the thymus in HIV-1 pathogenesis remains unclear. We developed an assay to quantify the number of recent thymic emigrants in blood based on the detection of a major excisional DNA byproduct (termed α1 circle) of T cell receptor rearrangement. By studying 532 normal individuals, we found that α1 circle numbers in blood remain high for the first 10–15 yr of life, a sharp drop is seen in the late teen years, and a gradual decline occurs thereafter. Compared with age-matched uninfected control individuals, α1 circle numbers in HIV-1–infected adults were significantly reduced; however, there were many individuals with normal α1 circle numbers. In 74 individuals receiving highly active antiretroviral therapy, we found no appreciable effect on α1 circle numbers in those whose baseline values were already within the normal range, but significant increases were observed in those with a preexisting impairment. The increases in α1 circle numbers were, however, numerically insufficient to account for the rise in levels of naive T lymphocytes. Overall, it is difficult to invoke thymic regenerative failure as a generalized mechanism for CD4 lymphocyte depletion in HIV-1 infection, as α1 circle numbers are normal in a substantial subset of HIV-1–infected individuals.
Article
Full-text available
Identifying the immunologic and virologic consequences of discontinuing antiretroviral therapy in HIV-infected patients is of major importance in developing long-term treatment strategies for patients with HIV-1 infection. We designed a trial to characterize these parameters after interruption of highly active antiretroviral therapy (HAART) in patients who had maintained prolonged viral suppression on antiretroviral drugs. Eighteen patients with CD4+ T cell counts ≥ 350 cells/μl and viral load below the limits of detection for ≥1 year while on HAART were enrolled prospectively in a trial in which HAART was discontinued. Twelve of these patients had received prior IL-2 therapy and had low frequencies of resting, latently infected CD4 cells. Viral load relapse to >50 copies/ml occurred in all 18 patients independent of prior IL-2 treatment, beginning most commonly during weeks 2–3 after cessation of HAART. The mean relapse rate constant was 0.45 (0.20 log10 copies) day−1, which was very similar to the mean viral clearance rate constant after drug resumption of 0.35 (0.15 log10 copies) day−1 (P = 0.28). One patient experienced a relapse delay to week 7. All patients except one experienced a relapse burden to >5,000 RNA copies/ml. Ex vivo labeling with BrdUrd showed that CD4 and CD8 cell turnover increased after withdrawal of HAART and correlated with viral load whereas lymphocyte turnover decreased after reinitiation of drug treatment. Virologic relapse occurs rapidly in patients who discontinue suppressive drug therapy, even in patients with a markedly diminished pool of resting, latently infected CD4+ T cells.
Article
Full-text available
The mechanism by which HIV-1 induces CD4(+) T cell death is not known. A fundamental issue is whether HIV-1 primarily induces direct killing of infected cells or indirectly causes death of uninfected bystander cells. This question was studied using a reporter virus system in which infected cells are marked with the cell surface protein placental alkaline phosphatase (PLAP). Infection by HIV-PLAP of peripheral blood mononuclear cells (PBMCs) and T cell lines leads to rapid depletion of CD4(+) T cells and induction of apoptosis. The great majority of HIV-induced T cell death in vitro involves direct loss of infected cells rather than indirect effects on uninfected bystander cells. Because of its proposed role in HIV-induced cell death, we also examined the Fas (CD95/Apo1) pathway in killing of T cells by HIV-1. Infected PBMCs or CEM cells display no increase in surface Fas relative to uninfected cells. In addition, HIV-1 kills CEM and Jurkat T cells in the presence of a caspase inhibitor that completely blocks Fas-mediated apoptosis. HIV-1 also depletes CD4+ T cells in PBMCs from patients who have a genetically defective Fas pathway. These results suggest that HIV-1 induces direct apoptosis of infected cells and kills T cells by a Fas-independent mechanism.
Article
To contribute to the understanding of the role of the thymus in humans in the reconstitution of naive (CD45RA+) T cells after bone marrow transplantation (BMT), we compared T-cell regeneration in a unique situation, namely a thymectomized cancer patient (15 years old), with that of thymus-bearing patients after allogeneic BMT. These cases shared features of transplantation (total body irradiation, HLA-matched donors, and graft-versus-host disease prophylaxis with cyclosporine A) and all had an uncomplicated posttransplantation course. As shown by fluorescence-activated cell sorting analyses, the thymectomized host failed to reconstitute CD45RA+ T-helper cells even 24 months after BMT (11% CD45RA+ of CD4+ cells). In this patient, preferentially CD45RO+ cells contributed to the recovery of CD4+ cells (206 of 261/μL at 6 months and 463 of 558/μL at 24 months after BMT, CD45RA+ of CD4+ cells), whereas CD45RA+ cells remained low (<60/μL). In contrast, nine thymus-bearing hosts (5 children and 4 adults) examined between 6 and 24 months after BMT effectively reconstituted CD4+/CD45RA+ cells according to their normal age-related range (≥28% in adults and ≥50% in children). Five of these were analyzed sequentially at 6 and 9 months after BMT. Within this period, CD45RA+ cells increasingly contributed to the recovery of CD4+ cells (median, +21%), even when total CD4+ cells decreased. With respect to T-cytotoxic/suppressor cells, the thymectomized host retained the capacity to recover CD45RA+ cells (137 of 333/μL at 6 months and 596 of 1,046/μL at 24 months after BMT, CD45RA+ of CD8+ cells), a proportion similar to that seen in thymus-bearing hosts. These findings suggest that a thymus-independent pathway exists to regenerate CD45RA+ T-cytotoxic/suppressor cells, but residual thymus is essential to reconstitute naive (CD45RA+) T-helper cells after BMT in humans.
Article
In human immunodeficiency virus (HIV)-1 infection, highly increased T-cell turnover was proposed to cause exhaustion of lymphocyte production and consequently development of AIDS. Here, we investigated cell proliferation, as measured by expression of the Ki-67 nuclear antigen, in peripheral blood CD4+ and CD8+ lymphocyte subpopulations before and during highly active antiretroviral therapy (HAART). In untreated HIV-1 infection, both the percentage and number of Ki-67+CD4+ and CD8+ lymphocytes were significantly increased, compared with values obtained from healthy individuals. A more than 10-fold increase in the percentage of dividing naive CD4+ T cells in the blood was found when the number of these cells were below 100 per μL.. HAART induced an immediate decline in Ki-67 antigen expression, despite often very low CD4+ T-cell numbers, arguing against increased proliferation being a homeostatic response. After approximately 24 weeks of HAART treatment, a transient increase in the number of proliferating cells was seen, but only in the CD4+CD27+ memory pool. In the CD8+ T-cell compartment, the number of dividing cells was elevated 20- to 25-fold. This increase was most notable in the CD27+ CD 45RO+ and CD27−CD45RO+ memory CD8+ T-cell pool, corresponding with the degree of expansion of these subsets. Reduction of plasma HIV-RNA load by HAART was accompanied by a decrease in numbers and percentages of dividing cells in all CD8+T-cell subsets. Taken together, our results indicate that peripheral T-cell proliferation is a consequence of generalized immune activation. (Blood. 2000;95:249-255)
Article
Hematopoietic stem cell transplantation (HSCT) is followed by profound immunodeficiency. Thymic function is necessary for de novo generation of T cells after HSCT. Circulating CD45RA+ naive T-cell levels are predictive of antigen-specific T-cell responses in the absence of graft-versus-host disease (GVHD). These T cells may not represent recent thymic emigrants, since naive T cells may maintain this phenotype if not antigen-activated. To accurately measure thymic output after HSCT and determine the factors that influence thymic function, T-cell receptor excision circles (TRECs) were examined in CD4+ and CD8+ cells from a cross-section of patients following HSCT. TREC levels rose weeks after HSCT and could be detected in patients 6 years after HSCT. TREC levels correlated with the frequency of phenotypically naive T cells, indicating that such cells were not expanded progeny of naive T cells present in the donor graft. Chronic GVHD was the most important factor that predicted low TREC levels even years after HSCT. Patients with a history of resolved GVHD had decreased numbers of TREC, compared with those with no GVHD. Because few adults had no history of GVHD, it was not possible to determine whether age alone inversely correlated with TREC levels. Recipients of cord blood grafts had no evidence of decreased TREC induced by immunosuppressive prophylaxis drugs. Compared with unrelated donor grafts, recipients of matched sibling grafts had higher TREC levels. Collectively, these data suggest that thymopoiesis is inhibited by GVHD. Larger studies will be needed to determine the independent contributions of age and preparative regimen to post-transplant thymopoietic capacity.
Article
Human immunodeficiency virus (HIV) infection is associated with progressive loss of circulating CD4+ lymphocytes. Treatment with highly active antiretroviral therapy (HAART) has led to increases in CD4+ T lymphocytes of naive (CD45RA+62L+) and memory (CD45R0+RA−) phenotypes. Thymic computerized tomography scans were obtained on 30 individuals with HIV disease to investigate the role of the thymus in cellular restoration after 48 weeks of HAART. Individuals with abundant thymic tissue had higher naive CD4+ T lymphocyte counts at weeks 2–24 after therapy than individuals with minimal thymic tissue. Individuals with abundant thymic tissue had significantly larger increases in naive CD4+ cells during the first 4 weeks of therapy. These individuals were also more likely to experience viral rebound despite comparable initial declines in plasma HIV-1 RNA. These findings suggest that there is a complex relationship among the thymus, viral replication, and lymphocyte restoration after application of HAART in HIV disease.
Article
In human immunodeficiency virus (HIV)-1 infection, highly increased T-cell turnover was proposed to cause exhaustion of lymphocyte production and consequently development of AIDS. Here, we investigated cell proliferation, as measured by expression of the Ki-67 nuclear antigen, in peripheral blood CD4(+) and CD8(+) lymphocyte subpopulations before and during highly active antiretroviral therapy (HAART). In untreated HIV-1 infection, both the percentage and number of Ki-67(+) CD4(+) and CD8(+) lymphocytes were significantly increased, compared with values obtained from healthy individuals. A more than 10-fold increase in the percentage of dividing naive CD4(+) T cells in the blood was found when the number of these cells were below 100 per microL. HAART induced an immediate decline in Ki-67 antigen expression, despite often very low CD4(+) T-cell numbers, arguing against increased proliferation being a homeostatic response. After approximately 24 weeks of HAART treatment, a transient increase in the number of proliferating cells was seen, but only in the CD4(+) CD27(+) memory pool. In the CD8(+) T-cell compartment, the number of dividing cells was elevated 20- to 25-fold. This increase was most notable in the CD27(+) CD 45RO(+) and CD27(-) CD45RO(+) memory CD8(+) T-cell pool, corresponding with the degree of expansion of these subsets. Reduction of plasma HIV-RNA load by HAART was accompanied by a decrease in numbers and percentages of dividing cells in all CD8(+) T-cell subsets. Taken together, our results indicate that peripheral T-cell proliferation is a consequence of generalized immune activation. (Blood. 2000;95:249-255)