Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2007 Dec;150(3):442–450. doi: 10.1111/j.1365-2249.2007.03526.x

Immune reconstitution in human immunodeficiency virus type 1-infected children with different virological responses to anti-retroviral therapy

A Anselmi *, D Vendrame *, O Rampon , C Giaquinto , M Zanchetta *, A De Rossi *
PMCID: PMC2219365  PMID: 17956580

Abstract

Immune repopulation, despite virological failure, often occurs in children under highly active anti-retroviral therapy (HAART). The aim of this study was to analyse the characteristics of immune repopulation and activation in children with and without virological response to HAART. Fourteen human immunodeficiency virus type 1 (HIV-1)-infected children with suppression of HIV-1 plasma viraemia (virological responders, VR) and 16 virological non-responders (VNR) to therapy were studied at baseline and after approximately 2 years of HAART. During therapy, CD4+ T cells increased in both groups, but were higher in the VR than in the VNR group. All CD4+ T cell subsets (naive, central memory, effector/memory and CD38+) increased significantly in VR children, while there was a significant increase only in naive cells in VNR children. Naive CD8+ T cells and T cell receptor rearrangement excision circles (TREC), an indicator of thymic output, increased in both VR and VNR children. Activated CD8+CD38+ T cells decreased in VR but remained high in VNR children. Levels of circulating lipopolysaccharide (LPS), an indicator of microbial translocation, further increased in VNR children. In conclusion, HAART induced an increase in naive cells in all children, regardless of their virological response. However, the persistence of viraemia resulted in an impaired expansion of memory CD4+ T cells susceptible to HIV-1 infection, and together with the microbial translocation sustained the persistence of a high level of immune activation.

Keywords: HAART, HIV-1-infected children, immune reconstitution, LPS, T cell activation

Introduction

Mother-to-child transmission of human immunodeficiency virus type 1 (HIV-1) accounts for most paediatric HIV-1 infections. In the absence of anti-retroviral therapy, the establishment of the viral set point in infants occurs slowly, suggesting that induction of anti-viral immunity is not as rapid as in adults [1, 2]. HIV-1 disease is characterized by chronic immune activation, and lymphocyte activation is a hallmark of disease progression [3]. Several studies suggest that, in addition to HIV-1 antigens, other viral and/or microbial antigens, proinflammatory cytokines and chemokines cause immune activation [46]. Increased translocation of luminal microbial products may also be involved [7]. Indeed, HIV-1 infection is localized predominantly to the gastrointestinal tract [8]; CD4+ T cell depletion and damage to the mucosal barrier lead to translocation of microbes and/or microbial products resulting in systemic immune activation [7].

The administration of highly active anti-retroviral therapy (HAART), which is usually a combination of protease and reverse transcriptase inhibitors, reduces HIV-1 load efficiently to undetectable levels and increases the number of circulating CD4+ T cells in children as well as adults [9, 10]. Although HAART does not eradicate HIV-1, due to a persistent low level of viral replication and the longevity of latently infected cells, it allows for substantial recovery of immune function [11, 12]. Immune reconstitution after HAART initiation is different in children. While, in the adult, it is a biphasic process with an initial rapid increase in memory CD4+ T cells followed by a slower and smaller increase in naive CD4+ T cells [13, 14], in children immune repopulation involves mainly naive cells, due probably to more efficient thymopoiesis [1518]. Furthermore, some children show a discordant response to HAART characterized by a significant increase in CD4+ T cell counts, despite persistent detectable viraemia [15, 16, 1922]. Although the mechanisms of this discordant response to HAART have not been elucidated fully, it has been suggested that thymic output [15, 16, 19, 23] and an impaired replicative capacity of drug-resistant viruses [20, 24, 25] might be involved.

How different CD4+ T cell subsets contribute to immune reconstitution and influence immune activation in virological responder and non-responder children remains to be clarified. The aim of this study was to analyse the characteristics of immune reconstitution and the status of immune activation in children with different virological responses to therapy.

Materials and methods

Patients

The study cohort consisted of 30 vertically HIV-1-infected children who were treated at the Pediatric Department of Padova University. The inclusion criterion was their response to HAART: 14 virological responder (VR) children had long-term viral load (VL) suppression (VL ≤ 400 copies/ml) and 16 virological non-responder (VNR) children had persistently detectable VL (VL > 400 copies/ml). All children were naive for protease inhibitor (PI) at HAART initiation. The HAART regimen consisted of a triple-drug combination, including two reverse transcriptase inhibitors (zidovudine, lamivudine, stavudine or nevirapine) and one protease inhibitor (nelfinavir, indinavir or ritonavir). Four VNR children enrolled in this study maintained a detectable VL despite changes in reverse transcriptase inhibitors. CD4+ and CD8+ T cell counts and plasma HIV-1 RNA levels were followed over time. Peripheral blood mononuclear cells (PBMC) were isolated from peripheral blood by centrifugation on Ficoll-Paque (Pharmacia, Uppsala, Sweden) gradient and then cryopreserved until use. Seventeen age-matched uninfected children, all born to HIV-1-seropositive mothers, were also analysed. The study was approved by the Institutional Ethical Committee; informed consent was obtained for patients and controls from parents or legal guardians.

Flow cytometry quantification of CD4+ and CD8+ T lymphocyte subpopulations

T lymphocyte subpopulations were stained for four-colour flow cytometry using the following labelled monoclonal antibodies (mAbs): anti-CD3 [fluorescein isothiocyanate (FITC)], anti-CD4 [peridinin chlorophyll protein (PerCP)], anti-CD8 (PerCP), anti-CD27 [phycoerythrin (PE)], anti-CD45RA [allophycocyanin (APC)] and anti-CD38 (PE). Appropriate isotypic controls (mouse IgG1-PE and mouse IgG2b-APC) were used to evaluate non-specific staining. mAbs were obtained from Becton-Dickinson (Becton-Dickinson Biosciences Pharmingen, San Diego, CA, USA). Acquisition was performed in a cytometer (FACSCalibur; Becton-Dickinson) cytometer using the CELLQuest Software (Becton-Dickinson). A total of 50 000 events were collected in the lymphocyte gate using morphological parameters (forward- and side-scatter). The percentage of CD45RA+, CD27+ and CD38+ cells was calculated within the CD3+CD4+ or CD3+CD8+ gate. The absolute cell count of different subsets was calculated by multiplying the percentage value by the total number of CD4+ or CD8+ T cells. CD45RA and CD27 expression was used to identify CD4+ and CD8+ T cell subsets as follows: CD45RA+CD27+ (naive), CD45RACD27+ (central memory), CD45RACD27 (effector/memory CD4+ and preterminally differentiated effector CD8+ T cells) and CD8+CD45RA+CD27 (terminally differentiated cytotoxic effector) [2630].

Quantification of T cell receptor rearrangement excision circles (TREC)

Thymic output was studied in six VR and eight VNR children by measuring TREC in sequential samples collected at HAART entry and during therapy (1, 3, 6, 9, 12, 18 and 24 months). TREC levels were analysed by real-time polymerase chain reaction (PCR), exactly as described previously [15, 16].

Lipopolysaccharide (LPS) levels

Plasma samples were diluted fivefold with endotoxin-free water and then heated to 70°C for 10 min to inactivate plasma proteins. Plasma LPS was quantified with a commercially available assay (Limulus amoebocyte lysate QCL-1000; Cambrex, Milan, Italy) according to the manufacturer's protocol.

Statistical analysis

Differences in characteristics among infants were analysed using non-parametric tests. The Mann–Whitney test was used to compare data between groups, and the Wilcoxon non-parametric test was used to compare data within a group. TREC and CD4+CD45RA+CD27+ T cell count data obtained in sequential samples were analysed by linear regression analysis, and the slopes of TREC/1 × 105 PBMC per day and CD4+CD45RA+CD27+ T cells/μl per day were calculated individually. Correlations between different parameters were analysed using the non-parametric Spearman's test. Analyses were performed with spss statistical software (version 12·0).

Results

Immunological and virological characteristics of HIV-1-infected children

Thirty HIV-1-infected children and 17 uninfected children were studied. All HIV-1-infected children were studied at baseline and after approximately 2 years (median 23 months; range 19–30 months) of HAART. The immunological and virological characteristics at baseline and after HAART are described in Table 1. Before therapy, HIV-1-infected children had a median plasma viraemia of 4·5 log10 copies/ml. Both CD4+ T cell percentage and count were significantly lower than those of HIV-1-uninfected controls. In contrast, the CD8+ T cell percentage was higher than that of uninfected children. At baseline, CD4+ and CD8+ T cell counts and percentages were not significantly different in the VR and VNR groups. After 2 years of therapy, there was a significant increase in CD4+ T cell percentage and count in both the VR (13–29%, P < 0·001, and 345–686 cells/μl, P < 0·001, respectively) and VNR groups (7–17%, P = 0·001, and 210–433 cells/μl, P = 0·008, respectively). After 2 years of HAART, CD8+ T cell percentage decreased in VR children (P = 0·003); however, the absolute CD8+ T cell counts did not change significantly in either group, yet tended to increase in VNR children.

Table 1.

Immunological and virological characteristics of the studied population of human immunodeficiency virus type 1 (HIV-1)-infected, highly active anti-retroviral therapy (HAART)-treated children.

Virological responder Virological non-responder


Characteristics Control group* Total number of HIV-1-infected children before HAART Pa Baseline After HAART Pb Baseline After HAART Pb
Number of children 17 30 14 14 16 16
Age (years) 8·7 9·3 9·4 11·8 6·4 8·4
(2·5–17) (2–14) (2·8–14·2) (4·8–16) (2–14) (4–17)
Lymphocyte subsets
 % CD4+ 28·8 10 <0·001 13·3 28·8 <0·001 7·4 17·6 0·001
(11–49) (0–34) (0–34) (18–49) (2·5–29) (6·5–35·7)
 CD4+/μl 750 272 <0·001 345 686 <0·001 210 433 0·008
(369–2338) (5–916) (5–571) (439–1851) (10–916) (63–1126)
 % CD8+ 17·5 44 <0·001 42·6 32·3 0·003 46 51 0·387
(9·9–29·1) (6–72) (25–72) (19–54) (20–58) (23–70)
 CD8+/μl 509 739 0·12 702 858 0·391 896 1078 0·332
(239–1884) (99–3664) (225–2046) (286–1570) (99–3664) (284–3451)
 CD4+/CD8+ 1·9 0·4 <0·001 0·4 1 <0·001 0·3 0·6 0·004
(1–3·1) (0·1–1·2) (0·1–1·2) (0·4–2) (0·1–0·9) (0·1–1·3)
HIV-1 RNA log10 copies/ml 4·5 4·3 1·6 <0·001 4·9 4·7 0·033
(2·6–6·1) (3·2–5·6) (1·6–2·4) (2·6–6·1) (2·9–5·6)
*

Control group: age-matched HIV-1-negative children. Values were expressed as the median (ranges in parenthesis). Pa values between the control group and total HIV-1-infected children at baseline. Pb values between baseline and after HAART in virological responders and virological non-responders.

T cell subsets in uninfected and HIV-1-infected children at baseline

At baseline, compared to the control group, HIV-1-infected children had significantly lower cell counts in all CD4+ T cell subsets (naive 157 versus 415 cells/μl, P < 0·0001; central memory 85 versus 290 cells/μl, P < 0·0001; effector/memory 25 versus 77 cells/μl, P = 0·0002) (Fig. 1a). Conversely, HIV-1-infected children had higher cell counts in all CD8+ T cell subsets (pre-effectors 131 versus 34 cells/μl, P = 0·002; cytotoxic effector 185 versus 37 cells/μl, P = 0·001; and central memory 204 versus 121 cells/μl, P = 0·038), except naive CD8+ T cells, which were significantly lower in HIV-1-infected than uninfected children (191 versus 316 cells/μl, P = 0·006) (Fig. 1b). Interestingly, CD4+CD38+ T cells were lower (65 versus 209 cells/μl, P < 0·0001), while CD8+CD38+ T cells were higher in infected versus uninfected children (116 versus 25 cells/μl, P = 0·0001) (Fig. 1b).

Fig. 1.

Fig. 1

The phenotypic characteristics of CD4+ and CD8+ T subsets from 17 human immunodeficiency virus type 1 (HIV-1)-uninfected children and 30 HIV-1-infected children at highly active anti-retroviral therapy initiation are illustrated. CD27, CD45RA and CD38 expression in CD3+CD4+ T cells (a) and CD3+CD8+ T cells (b) were analysed by flow cytometry. Boxes and whiskers represent the 25th−75th and 10th−90th percentiles, respectively; the median is the central line in each box. P-values were determined by the Mann–Whitney U-test.

Changes in T cell subsets during HAART in VR and VNR children

At baseline, CD4+ and CD8+ T cell subsets of VR children did not differ significantly from those of the VNR group, even if pre-effector (CD45RACD27), effector (CD45RA+CD27) and CD38+ CD8+ T cell counts were more variable in VNR than VR children (Fig. 2a,b). After 2 years of HAART, CD4+ T cell counts increased in both groups, but in a different manner. In VR children, the peripheral CD4+ T cell increase was significant in naive (168–462 cells/μl, P = 0·001), central memory (97–213 cells/μl, P = 0·002) and effector/memory cell subsets (32–59 cells/μl, P = 0·035) and reached values similar to those observed in the uninfected children (Fig. 3a). Conversely, in VNR children, only naive CD4+ T cells increased significantly (113–263 cells/μl, P = 0·015), while values of all other CD4+ T cell subsets remained significantly lower than those observed in uninfected children (Fig. 3b). CD4+CD38+ T cells increased in both VR and VNR groups (66–142 cells/μl, P = 0·002, and 65–124 cells/μl, P = 0·045, respectively; Fig. 3a,b). A positive correlation between CD4+CD38+ and CD4+CD45RA+CD27+ T cell counts was observed at baseline and after HAART in both VR (at baseline rs = 0·84, P = 0·0002; and after HAART rs = 0·55, P = 0·05) and VNR children (rs = 0·91, P < 0·0001; and rs = 0·80, P = 0·0002, respectively). There was also a positive correlation between the increase of naive and CD38+ CD4+ T cell counts in both VR (rs = 0·51, P = 0·04) and VNR groups (rs = 0·96, P < 0·0001). Although the total CD8+ T cell count did not change during HAART (Table 1), a rise in naive CD8+ T cell subsets was observed in both VR (from 195 to 358 cells/μl, P = 0·006) and VNR children (from 165 to 288 cells/μl, P = 0·004). No significant changes were observed in the other CD8+ T cell subsets (central memory, pre-effector and cytotoxic effector cells) in either group, although the absolute cell numbers tended to increase in VNR children. CD8+CD38+ T cells decreased significantly only in VR children (107–60 cells/μl, P = 0·003), while these remained high in VNR children (Fig. 3c,d).

Fig. 2.

Fig. 2

T cell subsets are illustrated in virological responder (VR) and virological non-responder (VNR) children at highly active anti-retroviral therapy initiation. CD27, CD45RA and CD38 expression in CD3+CD4+ T cells (a) and CD3+CD8+ T cells (b) were analysed in 14 VR and 16 VNR children by flow cytometry. Boxes and whiskers represent the 25th−75th and 10th−90th percentiles, respectively; the median is the central line in each box. P-values were determined by the Mann–Whitney U-test.

Fig. 3.

Fig. 3

T cell repopulation in virological responder (VR) and virological non-responder (VNR) children is illustrated. CD27, CD45RA and CD38 expression in CD3+CD4+ T cells (a and b) and CD3+CD8+ T cells (c and d) were analysed in 14 VR children (a and c) and in 16 VNR children (b and d) at baseline and after 2 years of highly active anti-retroviral therapy. Data were compared with those obtained from 17 uninfected children. Boxes and whiskers represent the 25th−75th and 10th−90th percentiles, respectively; the median is the central line in each box. P-values were determined by the Wilcoxon test.

Relationship between TREC and naive CD4+ T cells in VR and VNR children

TREC levels were quantified in six VR and eight VNR children for whom sequential samples during therapy were available. At baseline, TREC levels in PBMC and CD4+CD45RA+CD27+ T cell counts in the VR children did not differ significantly from those in the VNR group (Fig. 4a,b). After 2 years of HAART, TREC levels increased significantly in both VR (1503–5018, P = 0·04) and VNR children (595–2117, P = 0·03, Fig. 4a). In this subset of children the increase in CD4+CD45RA+CD27+ T cells occurred in both groups (Fig. 4b), and reflected the increase observed in the entire VR and VNR study groups (Fig. 3a,b). A positive correlation between TREC levels and CD4+CD45RA+CD27+ T cells was observed in both groups at baseline (VR, rs = 0·94, P = 0·005; VNR, rs = 0·83, P = 0·01) and after HAART (VR, rs = 0·88, P = 0·0019; VNR, rs = 0·81, P = 0·015). Although the slope of the increase in TREC did not differ significantly in VR and VNR children, the slope of the increase in CD4+ naive T cells was lower in VNR than VR children (Fig. 4c,d). Moreover, the slopes of the TREC were correlated positively with the slopes of the CD4+ naive T cells in VR (rs = 0·90, P = 0·037), but not in VNR children (rs = 0·48, P = 0·33).

Fig. 4.

Fig. 4

The dynamics of thymic output and CD4+CD45RA+CD27+ T cells in six virological responder (VR) and eight virological non-responder (VNR) children is illustrated. T cell receptor rearrangement excision circles (TREC) (a) and CD4+CD45RA+CD27+ T cell levels (b) were analysed at baseline and after 2 years of highly active anti-retroviral therapy. Slopes of TREC (c) and CD4+CD45RA+CD27+ T cells (d) in VR and VNR children. Boxes and whiskers represent the 25th−75th and 10th−90th percentiles, respectively; the central line in each box indicates the median. P-values were determined by the Wilcoxon (a and b) and the Mann–Whitney U-tests (c and d).

LPS levels in HIV-1-infected children

Plasma LPS levels were quantified in uninfected and HIV-1-infected children at baseline, before initiation of HAART. The patients showed significantly higher values than the control group (P = 0·0001; Fig. 5a); however, baseline LPS levels were not considerably different in the VR and VNR groups. After 2 years of HAART (Fig. 5b), LPS levels were unchanged in VR children but were increased still further in VNR children (56–67 pg/ml, P = 0·035).

Fig. 5.

Fig. 5

Microbial translocation in human immunodeficiency virus type 1 (HIV-1)-infected children is illustrated. Plasma lipopolysaccharide levels were determined in HIV-1-uninfected and HIV-1-infected children before highly active anti-retroviral therapy (HAART) initiation (a) and in virological responder (VR) and virological non-responder (VNR) children at baseline and after HAART (b). Boxes and whiskers represent the 25th−75th and 10th−90th percentiles, respectively; the central line in each box indicates the median. *P < 0·001 in comparison of the control group versus HIV-1-infected children.

Discussion

HAART has dramatically modified the course of HIV-1 infection, resulting in a drastic decrease of viral load to undetectable levels and a significant increase in the peripheral CD4+ T cell repopulation. In HAART-treated children, immune repopulation may occur despite persistent viraemia [15, 16, 1922]. To add to the scarce information available on immune reconstitution in HIV-1-infected children, we investigated peripheral immune repopulation in children with and without a virological response to therapy.

Our study demonstrated that immune reconstitution was quite different in VR and VNR children with regard to both CD4+ and CD8+ T cell compartments. Moreover, immune activation decreased in VR children, yet persisted and even increased in VNR children.

Prior to HAART initiation, VR and VNR patients had similar levels of plasma viraemia and did not show significant differences in any T cell subsets and TREC level. When compared to age-matched HIV-1-uninfected children, both VR and VNR children showed cell depletion in all CD4+ T cell subsets, but a higher cell count in all CD8+ T cell subsets except naive cells. This finding, together with the high level of activated CD8+CD38+ cells, is in agreement with the concept that HIV-1 infection drives a broad activation and differentiation of CD8+ T cells, pushing them from naive to late-stage differentiation [31].

After 2 years of HAART, immune reconstitution in VR children occurred in all cell subsets, while in the VNR children only naive CD4+ T cells increased. Of interest, both VR and VNR children showed an increase in TREC levels. This result is in agreement with previous findings [15, 16, 19, 21] and indicates that the persistence of viraemia during HAART does not impair the increase in thymic function and the subsequent output of naive cells. The finding that naive CD8+ T cells increased in both VR and VNR groups confirms that thymic function was recovered. However, the increase in TREC correlated with the increase in naive CD4+ T cells in VR, but not in VNR children, in agreement with a previous study [15]. In addition, for a similar TREC increase, the naive CD4+ T cell increase was lower in VNR than VR children. It is possible that thymic emigrants in the context of ongoing viraemia are activated from circulating viral antigen, proliferate and differentiate into memory cell subsets [32], which are more susceptible to HIV-1 infection than naive cells. Although the mechanisms underlying partial recovery of immune function despite virological failure remain to be elucidated, it is likely that HAART prevents HIV-1 replication in T cell progenitors [3335]. In addition, the impaired replicative capacity of drug-resistant viruses and the shift of viral co-receptor usage from CXCR4 expressed on naive cells to CCR5 expressed on memory CD4+ T cells [20, 33, 36] probably both contribute to peripheral immune repopulation by naive CD4+ T cells.

Along with the rise in CD4+, CD4+CD38+ cells increased in both VR and VNR groups and these were correlated positively with increases in naive cells. As opposed to what occurs in adults, these results suggest that CD38 is a marker of cell immaturity rather than cell activation in CD4+ T cells in children [37, 38]. Finding that its expression is high in naive CD4+ T cells [39], that it decreases over time [40] and that it increases in CD4+ T cells during immune reconstitution [37, 38] all support this concept. Besides representing a marker of cell immaturity, CD38 in CD4+ T cells is likely to have multiple functional activities [37]. Further studies are required to clarify in depth the role of CD38 in CD4+ T cells in children.

None the less, our study demonstrates that persistence of viraemia greatly impairs the expansion of memory and effector CD4+ T cell subsets. In VR children, these cell subsets increased and reached levels similar to those observed in uninfected controls. The peripheral CD4+ T cell subset repopulation may be due to an expansion of memory and effector CD4+ T cells, cellular redistribution from tissue/lymph nodes to the periphery and reduced apoptosis [41]. In VNR children, these cell subsets remained significantly lower. This may be due to the persistence of circulating virions in VNR children and also to the greater susceptibility of these cells to HIV-1 infection compared to naive cells [33, 42]. The peripheral depletion may also be induced by a decrease in cell proliferation and an increase in cell death [41]. Further studies with markers of cell apoptosis and survival, not feasible in the present analysis due to the small numbers of cells available, will be very useful to clarify this aspect.

With regard to the CD8+ T cell compartment, the percentage of CD8+ T cells decreased in VR children; this percentage decrease was significant not only because of the CD8+ lowering effect, but also because CD4+ increased notably. No significant changes occurred in memory and effector cell subsets, yet absolute cell counts tended to further increase in VNR children. This finding suggests that HIV-1-driven chronic stimulation and activation continued to persist in VNR children. This was also supported by the finding that activated CD8+CD38+ T cells decreased significantly in VR children, yet remained persistently high in VNR children. Reduction of CD38 expression in the VR group may reflect a reduction of antigenic stimulation due to the undetectable plasma viraemia [37, 43]. Immune activation is driven not only by HIV-1, but also by other pathogens. HIV-1 infection occurs in the gastrointestinal tract, damaging the immune component of the gastrointestinal mucosa, with subsequent translocation of microbial products such as LPS, a potent immunostimulatory molecule that affects systemic immune activation. At baseline, circulating levels of LPS were higher in HIV-1-infected children than in controls. While a reduction in LPS levels has been reported in HIV-1-infected adults during therapy [7], LPS levels remained roughly stable in VR children, but tended to increase even further in VNR children during HAART. This finding strongly supports the concept that persistence of immune activation in children might be caused not only by persistence of HIV-1 antigens but also by other pathogens and microbial products.

In summary, this study has demonstrated that HAART induces immune repopulation of different CD4+ T cell subpopulations only in children with a virological response to therapy. Moreover, persistence of viraemia did not affect thymic output and the increase in naive cells, but greatly impaired CD4+ T cell differentiation. The persistence of HIV-1 virions/antigens, together with circulating microbial products, sustained high levels of activated CD8+CD38+ T cells. The persistence of systemic immune activation might ultimately determine the progression to AIDS.

Acknowledgments

We thank Pierantonio Gallo for artwork and Lisa Smith for editorial assistance. This work was supported by ISS grant no. 45G.12, grant no. 40 G21 and by the PENTA Foundation.

References

  • 1.De Rossi A, Masiero S, Giaquinto C, et al. Dynamics of viral replication in infants with vertically acquired human immunodeficiency virus type 1 infection. J Clin Invest. 1996;97:323–30. doi: 10.1172/JCI118419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Luzuriaga K, Wu H, McManus M, et al. Dynamics of human immunodeficiency virus type 1 replication in vertically infected infants. J Virol. 1999;73:362–7. doi: 10.1128/jvi.73.1.362-367.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Giorgi JV, Hultin LE, McKeating JA, et al. Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage. J Infect Dis. 1999;179:859–70. doi: 10.1086/314660. [DOI] [PubMed] [Google Scholar]
  • 4.Sankaran S, Guadalupe M, Reay E, et al. Gut mucosal T cell responses and gene expression correlate with protection against disease in long-term HIV-1-infected nonprogressors. Proc Natl Acad Sci USA. 2005;102:9860–5. doi: 10.1073/pnas.0503463102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Valdez H, Lederman MM. Cytokines and cytokine therapies in HIV infection. AIDS Clin Rev. 1997:187–228. 1998. [PubMed] [Google Scholar]
  • 6.Behbahani H, Walther-Jallow L, Klareskog E, et al. Proinflammatory and type 1 cytokine expression in cervical mucosa during HIV-1 and human papillomavirus infection. J Acquir Immune Defic Syndr. 2007;45:9–19. doi: 10.1097/QAI.0b013e3180415da7. [DOI] [PubMed] [Google Scholar]
  • 7.Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–71. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 8.Mehandru S, Poles MA, Tenner-Racz K, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200:761–70. doi: 10.1084/jem.20041196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Purswani M, Johann-Liang R, Cervia J, Noel GJ. Effect of changing antiretroviral therapy on human immunodeficiency virus viral load: experience with fifty-four perinatally infected children. Pediatr Infect Dis J. 1999;18:512–6. doi: 10.1097/00006454-199906000-00007. [DOI] [PubMed] [Google Scholar]
  • 10.Gibb DM, Newberry A, Klein N, De Rossi A, Grosch-Woerner I, Babiker A. Immune repopulation after HAART in previously untreated HIV-1-infected children. Lancet. 2000;355:1331–2. doi: 10.1016/s0140-6736(00)02117-6. Paediatric European Network for Treatment of AIDS (PENTA) Steering Committee. [DOI] [PubMed] [Google Scholar]
  • 11.Picker LJ, Maino VC. The CD4+ T cell response to HIV-1. Curr Opin Immunol. 2000;12:381–6. doi: 10.1016/s0952-7915(00)00104-7. [DOI] [PubMed] [Google Scholar]
  • 12.Lederman MM, Valdez H. Immune restoration with antiretroviral therapies. JAMA. 2000;284:223–8. doi: 10.1001/jama.284.2.223. [DOI] [PubMed] [Google Scholar]
  • 13.Pakker NG, Notermans DW, De Boer RJ, et al. Biphasic kinetics of peripheral blood T cells after triple combination therapy in HIV-1 infection: a composite of redistribution and proliferation. Nat Med. 1998;4:208–14. doi: 10.1038/nm0298-208. [DOI] [PubMed] [Google Scholar]
  • 14.Kaufmann GR, Zaunders J, Murray J, et al. Relative significance of different pathways of immune reconstitution in HIV type 1 infection as estimated by mathematical modeling. AIDS Res Hum Retroviruses. 2001;17:147–59. doi: 10.1089/08892220150217238. [DOI] [PubMed] [Google Scholar]
  • 15.Ometto L, De Forni D, Patiri F, et al. Immune reconstitution in HIV-1-infected children on antiretroviral therapy: role of thymic output and viral fitness. AIDS. 2002;16:839–49. doi: 10.1097/00002030-200204120-00003. [DOI] [PubMed] [Google Scholar]
  • 16.De Rossi A, Walker AS, Klein N, De Forni D, King D, Gibb DM. Increased thimic output after initiation of antiretroviral therapy in human immunodeficiency virus type 1-infected children in the Paediatric European Network for treatment of AIDS (PENTA) 5 trial. J Infect Dis. 2002;186:312–20. doi: 10.1086/341657. [DOI] [PubMed] [Google Scholar]
  • 17.Resino S, Galan I, Bellon JM, Navarro ML, Leon JA, Munoz-Fernandez MA. Characterizing the immune system after long-term undetectable viral load in HIV-1-infected children. J Clin Immunol. 2003;23:279–89. doi: 10.1023/a:1024536816684. [DOI] [PubMed] [Google Scholar]
  • 18.Fraaij PL, Verweel G, Van Rossum AM, et al. Sustained viral suppression and immune recovery in HIV type 1-infected children after 4 years of highly active antiretroviral therapy. Clin Infect Dis. 2005;40:604–8. doi: 10.1086/427696. [DOI] [PubMed] [Google Scholar]
  • 19.Chavan S, Bennuri B, Kharbanda M, Chandrasekaran A, Bakshi S, Pahwa S. Evaluation of T cell receptor gene rearrangement excision circles after antiretroviral therapy in children infected with human immunodeficiency virus. J Infect Dis. 2001;183:1445–54. doi: 10.1086/320197. [DOI] [PubMed] [Google Scholar]
  • 20.Bagnarelli P, Vecchi M, Burighel N, et al. Genotypic and phenotypic correlates of the HIV type 1 env gene evolution in infected children with discordant response to antiretroviral therapy. AIDS Res Hum Retroviruses. 2004;20:1306–13. doi: 10.1089/aid.2004.20.1306. [DOI] [PubMed] [Google Scholar]
  • 21.Resino S, Galan I, Perez A, et al. HIV-infected children with moderate/severe immune-suppression: changes in the immune system after highly active antiretroviral therapy. Clin Exp Immunol. 2004;137:570–7. doi: 10.1111/j.1365-2249.2004.02583.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ghaffari G, Passalacqua DJ, Caicedo JL, Goodenow MM, Sleasman JW. Two-year clinical and immune outcomes in human immunodeficiency virus-infected children who reconstitute CD4 T cells without control of viral replication after combination antiretroviral therapy. Pediatrics. 2004;114:604–11. doi: 10.1542/peds.2004-0274. [DOI] [PubMed] [Google Scholar]
  • 23.Lecossier D, Bouchennet P, Schneider P, Clavel F, Hance AJ. Discordant increases in CD4+ T cells in human immunodeficiency virus-infected patients experiencing virologic treatment failure. Role of changes in thymic output and T cell death. J Infect Dis. 2001;183:1009–16. doi: 10.1086/319285. [DOI] [PubMed] [Google Scholar]
  • 24.Stoddart CA, Liegler TJ, Mammano F, et al. Impaired replication of protease inhibitor-resistant HIV-1 in human thymus. Nat Med. 2001;7:712–8. doi: 10.1038/89090. [DOI] [PubMed] [Google Scholar]
  • 25.Hunt P, Deeks S, Bangsberg D, et al. The independent effect of drug resistance on T cell activation in HIV infection. AIDS. 2006;20:691–9. doi: 10.1097/01.aids.0000216369.30948.18. [DOI] [PubMed] [Google Scholar]
  • 26.Van Baarle D, Kostense S, Van Oers MH, Hamann D, Miedema F. Failing immune control as a result of impaired CD8+ T-cell maturation: CD27 might provide a clue. Trends Immunol. 2002;23:586–91. doi: 10.1016/s1471-4906(02)02326-8. [DOI] [PubMed] [Google Scholar]
  • 27.Schiott A, Lindstedt M, Johansson-Lindbom B, et al. CD27− CD4+ memory T cells define a differentiated memory population at both the functional and transcriptional levels. Immunology. 2004;113:363–70. doi: 10.1111/j.1365-2567.2004.01974.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bekker V, Bronke C, Scherpbier HJ, et al. Cytomegalovirus rather than HIV triggers the outgrowth of effector CD8+CD45RA:+CD27− T cells in HIV-1-infected children. AIDS. 2005;19:1025–34. doi: 10.1097/01.aids.0000174448.25132.ad. [DOI] [PubMed] [Google Scholar]
  • 29.Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–63. doi: 10.1146/annurev.immunol.22.012703.104702. [DOI] [PubMed] [Google Scholar]
  • 30.Emu B, Sinclair E, Favre D, et al. Phenotypic, functional, and kinetic parameters associated with apparent T cell control of human immunodeficiency virus replication in individuals with and without antiretroviral treatment. J Virol. 2005;79:14169–78. doi: 10.1128/JVI.79.22.14169-14178.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Jordan KA, Furlan SN, Gonzalez VD, et al. CD8 T cell effector maturation in HIV-1-infected children. Virology. 2006;347:117–26. doi: 10.1016/j.virol.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 32.Brenchley JM, Douek DC, Ambrozak DR, et al. Expansion of activated human naive T-cells precedes effector function. Clin Exp Immunol. 2002;130:432–40. doi: 10.1046/j.1365-2249.2002.02015.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang YM, Wang B, Dyer WB, Lachireddy K, Peng NK, Saksena NK. Evidence for predominance of CCR5-using HIV-1 strains during highly active antiretroviral therapy. Curr HIV Res. 2007;5:221–34. doi: 10.2174/157016207780077048. [DOI] [PubMed] [Google Scholar]
  • 34.Brenchley JM, Hill BJ, Ambrozak DR, et al. T-cell subsets that harbor human immunodeficiency virus (HIV) in vivo: implications for HIV pathogenesis. J Virol. 2004;78:1160–8. doi: 10.1128/JVI.78.3.1160-1168.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ye P, Kirschner DE, Kourtis AP. The thymus during HIV disease: role in pathogenesis and in immune recovery. Curr HIV Res. 2004;2:177–83. doi: 10.2174/1570162043484898. [DOI] [PubMed] [Google Scholar]
  • 36.Mammano F, Trouplin V, Zennou V, Clavel F. Retracing the evolutionary pathways of human immunodeficiency virus type 1 resistance to protease inhibitors: virus fitness in the absence and in the presence of drug. J Virol. 2000;74:8524–31. doi: 10.1128/jvi.74.18.8524-8531.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Savarino A, Bottarel F, Malavasi F, Dianzani U. Role of CD38in HIV-1 infection: an epiphenomenon of T-cell activation or an active player in virus/host interactions? AIDS. 2000;14:1079–89. doi: 10.1097/00002030-200006160-00004. [DOI] [PubMed] [Google Scholar]
  • 38.Resino S, Seoane E, Gutierrez MD, León JA, Muñoz-Fernández MA. CD4+ T-cell immunodeficiency is more dependent on immune activation than viral load in HIV-infected children on highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2006;42:269–76. doi: 10.1097/01.qai.0000222287.90201.d7. [DOI] [PubMed] [Google Scholar]
  • 39.De Martino M, Rossi ME, Azzari C, Gelli MG, Galli L, Vierucci A. Different meaning of CD38 molecule expression on CD4+ and CD8+ cells of children perinatally infected with human immunodeficiency virus type 1 infection surviving longer than five years. Pediatr Res. 1998;43:752–8. doi: 10.1203/00006450-199806000-00007. [DOI] [PubMed] [Google Scholar]
  • 40.De Vries E, De Bruin-Versteeg S, Comans-Bitter WM, et al. Longitudinal survey of lymphocyte subpopulations in the first year of life. Pediatr Res. 2000;47:528–37. doi: 10.1203/00006450-200004000-00019. [DOI] [PubMed] [Google Scholar]
  • 41.McCune JM. The dynamics of CD4+ T-cell depletion in HIV disease. Nature. 2001;410:974–9. doi: 10.1038/35073648. [DOI] [PubMed] [Google Scholar]
  • 42.Douek DC, Brenchley JM, Betts MR, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature. 2002;417:95–8. doi: 10.1038/417095a. [DOI] [PubMed] [Google Scholar]
  • 43.Rosenblatt H, Stanley K, Song L, et al. Immunological response to highly active antiretroviral therapy in children with clinically stable HIV-1 infection. J Infect Dis. 2005;192:445–55. doi: 10.1086/431597. [DOI] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES