Impaired telomerase activity in uninfected haematopoietic... : AIDS (original) (raw)

Introduction

The occurrence of peripheral blood cytopenia (thrombocytopenia, granulocytopenia, anaemia) and various dysplastic lesions of the bone marrow, which are often associated with HIV-1 infection, together with in vitro impaired survival/proliferation, with or without differentiation capacity of CD34+ haematopoietic progenitor cells (HPC) isolated from HIV-1-infected patients, indicates the presence of a virus-dependent lesion of the HPC compartment. However, the bulk of the available experimental evidence points to the lack of HIV-1 susceptibility of CD34+ HPC, and the virusdependent suppressive effect does not seem to require an HPC active or latent HIV-1 infection [1,2].

Although the mechanism responsible for the HIV-1-induced lesion of the HPC compartment may be multifactorial, a major pathogenetic role appears to be played by the apoptosis of CD34+ HPC, triggered by the interaction of free or virus-associated HIV-1 gp120 glycoprotein with the cell membrane of uninfected CD34+ HPC [3–6], a large subset of which express low amounts of CD4 molecules [7,8]. This event may in turn be associated with increased production of endogenous transforming growth factor (TGF)-β1 [9] through upregulation of TGF-β1 promoter activity [10]. TGF-β1 is a cell cycle blocker that acts on target cells by arresting or delaying cells in the G0/G1 phase of the cell cycle [11,12], as well as by inducing apoptosis [13].

TGF-β1 has been shown to inhibit telomerase activity in tumour cells [14]. Telomeres are nucleoprotein complexes consisting of tandem arrays of TTAGGG repeats, which are bound to specific proteins and form the end of eukaryotic chromosomes [15]. In normal human cells, telomeres shorten with successive cell divisions and the loss of telomeric DNA may serve as a mitotic clock that signals replicative cell senescence, exit from cell cycle and programmed cell death (apoptosis). Telomerase is required to maintain telomere lengths. Telomerase, an RNA-protein complex, is a reverse transcriptase (RT) that uses its RNA component to specify the addition of telomeric repeats to chromosome ends [16]. In humans, telomerase is expressed only in cells with essentially unlimited replicative potential, such as reproductive cells in ovaries and testes, and is repressed in most somatic cells and tissues [16,17]. Telomerase is highly expressed in cancer tissues [16,17], although, at least in mice, its presence is not required for oncogenic transformation or tumour formation [18].

The ageing of long-term self-renewing haematopoietic stem/progenitor cells is a process still incompletely known [19]. The recent observation that human HPC express telomerase activity [20,21] suggests a possible mechanism by which their ageing process might be delayed, if telomere length is indeed a mitotic clock regulating replicative potential.

We therefore compared the level of telomerase activity expressed in CD34+ HPC isolated from HIV-1-infected patients with the enzyme levels detectable in the same cell types isolated from normal donors. The influence of in vitro exposure to gp120 or TGF-β1 on telomerase levels expressed by normal human CD34+ HPC was also investigated.

Materials and methods

Study population

We studied 11 HIV-1-seropositive (nine men and two women, aged 24–35 years) and 15 HIV-1-seronegative (12 men and three women, aged 26–49 years) subjects, who gave their informed consent according to the 1975 Helsinki declaration. Nine peripheral blood and six bone-marrow samples were obtained from healthy donors. For ethical and clinical reasons, only peripheral blood samples were obtained from the 11 HIV-1-seropositive patients studied. However, circulating HPC are considered more ancestral than their bonemarrow counterparts [22,23].

At the time of sampling, both HIV-1 p24 core antigen and HIV-1 RNA loads were quantified in peripheral blood plasma specimens from all HIV-1-seropositive patients. HIV-1 p24 antigenaemia, after immune complex dissociation with basic treatment, was evaluated using a commercial kit (Vironostika HIV-1 Antigen Microelisa System, Organon Teknika, Durham, North Carolina, USA), and HIV-1 RNA quantification was performed using a commercial isothermal amplification kit (NucliSense, Organon Teknika), according to the manufacturer's instructions.

Isolation of CD34+ HPC

CD34+ HPC were isolated using the CD34 Progenitor Cell Isolation Kit (Miltenyi Biotec, Bergisch Gladbach, Germany). Briefly, light density mononuclear cells were initially isolated by density gradient centrifugation (density, 1.077 g/ml; Ficoll-Histopaque, Pharmacia, Uppsala, Sweden). CD34+ cells were then positively selected from the mononuclear cell population by immunomagnetic activated cell sorting [24] using the QBEND/10 antiCD34 monoclonal antibody (MAb; Miltenyi Biotech). Isolated CD34+ cells were more than 95% pure, as determined from their positive reactivity to immunofluorescence staining with an anti-CD34 MAb recognizing a different epitope (HPCA-2; Becton Dickinson, Mountain View, California, USA). Purified CD34+ HPC were either immediately assayed for telomerase activity, or pretreated with gp120 or TGF-β1.

The presence of HIV-1 proviral DNA in CD34+ cells purified from HIV-1-seropositive subjects was examined by PCR in samples of 20 000 CD34+ cells amplified using HIV-1 _gag_-specific primers SK38/SK39, following a previously described procedure [3] with a sensitivity of 10 proviral copies in a background of 104 cells. Positive controls were represented by H9 and Jurkat T-cell lines chronically infected with HIV-1, and negative controls were represented by PCR runs including all reagents except DNA.

Telomerase assay

Telomerase activity was measured by a PCR-based telomeric repeat amplification protocol (TRAP) [25,26] using the TRAPeze Telomerase Detection Kit (Oncor, Inc., Gaithersburg, Maryland, USA). Briefly, 3-[(3-cholamidopropyl)-ethylammonio]-1-propane-sulfonate (CHAPS) detergent lysis buffer extracts were prepared from isolated bone-marrow and peripheral blood CD34+ HPC. Protein concentrations were determined using the Bradford assay (Bio-Rad Laboratories, Milan, Italy) with bovine serum albumin as a standard, and 0.3 µg proteins were added to each reaction mixture [26] containing a TS (telomerase substrate) primer which was [γ32P]-labelled at the 5′-end using [γ32P]-ATP (Amersham, Milan, Italy) and T4 polynucleotide kinase (Pharmacia Biotech, Uppsala, Sweden), a mixture of TRAP primers, and Taq DNA polymerase in a final volume of 50 µl.

The reaction mixture was maintained at 30°C for 30 min to allow the telomerase to catalyse the addition of a number of telomeric repeats (CCTTAG) onto the 3′-end of the oligonucleotide substrate (TS) and then was immediately transferred at 80°C to inactivate telomerase. The extended products were then amplified by 25 cycles of PCR, with a 30 sec denaturation step at 94°C, 30 sec annealing step at 50°C, and 45 sec extension step at 72°C, using a 9600 Perkin Elmer thermocycler (Perkin Elmer, Monza, Italy). The amplified products (a ladder of products with six base increments, starting at 50 nucleotides) were resolved by polyacrylamide (9%) gel electrophoresis, run at 2500 V for 2 h at 50°C using a Genom LR instrument (Beckman Analytical, Milan, Italy). Finally, gels were dried and the TRAP products visualized by autoradiography (Hyperfilm-MP, Amersham). Positive telomerase controls consisted of human epithelioid tumour HeLa cell line extracts [Cell Bank, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia (IZSLE), Brescia, Italy], whereas negative controls consisted of human embryo diploid pulmonary fibroblast PEU cell strain extracts (Cell Bank, IZSLE), and CD34+ HPC extracts were pretreated with RNase A (Pharmacia Biotech) used at 5 µg/0.3 µg of proteins for 1 h at 37°C. PCR amplification controls included internal (TRAPeze kit) control oligonucleotides K1 and TSK1, which, together with TS, produced a 36 base-pair band in every lane in order to rule out the presence of Taq DNA polymerase inhibitors in cell extracts, and TRAP assays performed with CHAPS lysis buffer substituted for cell extracts.

TRAP reaction were quantified as follows: the autoradiographic signal intensity of each band of the TRAP product ladders was measured individually using an image analyser (UVP Image Store 7500, Ultra Violet Products Ltd, Cambridge, UK) in combination with Gel Works 1D Gel Analysis software (Ultra Violet Products Ltd) and corrected for background levels. The corrected signals of the products in each lane were then summed to yield arbitrary units of activity. The relative specific telomerase activity in each cell extract was expressed as a percentage of the specific activity obtained with HeLa cell extracts run in the same experiments. All experiments were performed in triplicate.

Telomerase assay in cells pretreated with gp120 or TGF-β1

In order to evaluate the influence of either HIV-1 envelope glycoprotein gp120 or TGF-β1 on the levels of telomerase expressed by HPC, different aliquots of 1 × 106 CD34+ HPC isolated from three bone-marrow and five peripheral blood samples from healthy HIV-1-seronegative donors were washed three times in Iscove's modified Dulbecco's medium (IMDM; Gibco, Grand Island, New York, USA) supplemented with 10% of fetal calf serum (FCS; Gibco), pelletted in 1.5 ml Eppendorf tubes and resuspended in 1 ml IMDM plus 10% FCS. Various aliquots of all cell suspensions were then added with 1 µg recombinant gp120 (rgp120; American Biotechnologies, Cambridge, Massachusetts, USA). In addition, different aliquots of the cell preparations obtained from peripheral blood samples were added with 10 ng recombinant TGF-β1 (rTGF-β1; R&D Systems, Inc., Minneapolis, Minnesota, USA). All cell preparations were incubated for 2 h at 37°C, and washed using low speed centrifugation, resuspended in 1 ml fresh growth medium supplemented with 1 ng/ml interleukin-3 (Genzyme, Cinisello Balsamo, Milan, Italy) and incubated at 37°C for up to 72 h. At the end of the incubation time, CD34+ HPC exposed to either rgp120 or rTGF-β1, together with duplicate mock-treated control cell cultures, were studied for the presence of telomerase activity, as described above. Purity and immunological specificity of rgp120 and rTGF-β1 were checked in our laboratory by Western blot assay using specific MAb (R&D Systems). Moreover, the commercial preparations of rgp120 and rTGF-β1 were found to be free of tumour necrosis factor (TNF)-α and TNF-β by immunoenzymatic assay (R&D Systems) and bacterial endotoxin (Limulus amoebocyte lysate test; Whittaker Bioproducts, Walkersville, Massachusetts, USA).

Cell viability assay

The viability of cells exposed to either rgp120 or rTGF-β1 was checked at the end of the incubation period by trypan blue dye exclusion assay. At the same time, cell extracts of four out of five samples isolated from peripheral blood of healthy donors were also analysed (using standard procedures, with an Olympus AU 5200 automatic analyser, Merck, Darmstadt, Germany) for levels of lactate dehydrogenase (LDH), glutamic oxaloacetic transaminase (GOT) and creatine kinase, in comparison with untreated control cell preparations.

Statistical analysis

The mean of the values ± SD for different experiments are shown. Significant differences between groups were determined using either the Mann-Whitney test, or the two-tailed Student's t test for paired samples.

Results

HIV-1-seropositive patient study population

Clinical and virological data characterizing the patients enrolled in the study are shown in Table 1. The patients were in different stages of disease, ranging from stages A1 to C3 of the Centers for Disease Control and Prevention classification [27]. All patients were viraemic, with an RNA viral load range of 100–33 000 HIV-1 RNA copies/ml plasma, with no relationship between RNA viral load and disease stage. Only three patients presented a detectable antigenaemia, with p24 antigen values ranging from 16 to 100 pg/ml. At the time of the study, only five out of 11 HIV-1-seropositive patients examined were under antiretroviral therapy with various combinations of nucleoside or non-nucleoside RT inhibitors or HIV-1 protease inhibitors. All patients had experienced one or more cycles of antiretroviral therapy with zidovudine.

T1-6

Table 1:

Stage of disease, viral markers and antiretroviral therapy in HIV-1-seropositive patients.

Telomerase activity in CD34+ HPC isolated from HIV-1-seropositive patients and healthy donors

All CD34+ cell populations isolated from HIV-1-seropositive patients were negative for the presence of HIV-1 proviral DNA (data not shown). All controls of telomerase assays gave the results that were expected. The results of some typical experiments for the determination of telomerase activity are shown in Fig. 1. The results obtained from all HPC populations studied are summarized in Table 2.

F1-6

Fig. 1:

. Results of telomeric repeat amplification protocol telomerase assays in CD34+ haematopoietic progenitor cells isolated from either the peripheral blood or bone marrow of healthy donors and the peripheral blood of HIV-1-infected patients. Lanes 1–3, CD34+ cells isolated from the bone marrow of healthy donors; lanes 4–6, CD34+ cells isolated from the peripheral blood of HIV-1-infected patients; lanes 7–9, CD34+ cells isolated from the peripheral blood of healthy donors; lane 10, HeLa cell line (positive control); lane 11, HeLa cell extract treated with RNase (negative control); lane 12, PEU cell strain (negative control).

T2-6

Table 2:

Telomerase activity in CD34+ haematopoietic progenitor cells (HPC) isolated from HIV-1-seropositive patients, in comparison with the telomerase activity expressed by CD34+ HPC isolated from healthy donors.

CD34+ HPC isolated from either peripheral blood or bone marrow of healthy donors expressed significant levels of telomerase activity, which were sometimes higher than those observed in HeLa cells used as positive controls (Table 2). Moreover, even if peripheral blood CD34+ HPC were considered more ancestral than those isolated from bone marrow [22,23], telomerase levels were apparently lower in peripheral blood than in bone-marrow CD34+ cells of healthy donors, although the difference did not reach statistical significance.

However, CD34+ HPC isolated from the peripheral blood of HIV-1-seropositive patients behaved in a significantly different way (P < 0.001). In fact, these cells did not express any detectable telomerase activity in nine patients, and showed a clearly reduced enzymatic activity in two patients. These results unequivocally demonstrated an inhibited telomerase activity in peripheral blood CD34+ HPC from HIV-1-infected patients that was irrespective of the disease stage, presence of p24 antigenaemia, plasma viral RNA load level, and concurrent antiretroviral therapy.

Telomerase assay in normal CD34+ HPC pretreated with gp120 or TGF-β1

The results of one experiment for telomerase assays in CD34+ HPC isolated from either peripheral blood or bone marrow from healthy donor subjects, maintained for 72 h in culture after exposure to rgp120 or rTGF-β1, are shown in Fig. 2. All the results of this set of experiments are summarized in Table 3.

F2-6

Fig. 2:

. Results of telomeric repeat amplification protocol telomerase assays in CD34+ haematopoietic progenitor cells isolated from either peripheral blood or bone marrow from healthy donors, 72 h after the exposure to either recombinant gp120 (rgp120) or recombinant transforming growth factor (rTGF)-β1. Lane 1, HeLa cell line (positive control); lane 2, HeLa cell extract treated with RNase (negative control); lane 3, PEU cell strain (negative control); lanes 4 and 5, CD34+ cells isolated from the peripheral blood of healthy donors treated with rgp120; lane 6, CD34+ cells isolated from the bone marrow of healthy donors treated with rgp120; lanes 7–9, CD34+ cells isolated from the peripheral blood of healthy donors treated with rTGF-β1; lanes 10 and 11, untreated CD34+ cells isolated from the peripheral blood of healthy donors; lane 12, untreated CD34+ cells isolated from the bone marrow of a healthy donor.

T3-6

Table 3:

Telomerase activity* in CD34+ haematopoietic progenitor cells (HPC) isolated from peripheral blood or bone marrow from healthy donor subjects 72 h after the exposure to recombinant gp120 (rgp120) or recombinant transforming growth factor (rTGF)- β1.

The results obtained showed that exposure to both rgp120 and rTGF-β1 specifically and significantly reduced telomerase activity with respect to untreated control cell preparations (P < 0.001). The inhibition of telomerase activity after exposure to rgp120 was significantly higher than that observed in the presence of rTGF-β1 (P < 0.05). Under these culture conditions, no significant loss of cell viability was observed at the end of the incubation period in cell preparations exposed to either rgp120 or rTGF-β1. In addition, all cell samples analysed for LDH, GOT and creatine kinase levels showed the same enzyme levels as those present in untreated control cell preparations (data not shown).

Discussion

Shortened telomeres have been demonstrated in T-lymphocyte subsets of HIV-1-infected patients, and the inferred impairment of their replicative potential has been claimed to be a possible mechanism for the decline of selected lymphocyte populations and the exhaustion of T-cell response [28–30].

Our study was the first to demonstrate severely impaired telomerase activity in CD34+ HPC isolated from HIV-1-infected patients in comparison with the same cell population types isolated from healthy donors. By employing the highly sensitive TRAP assay, all CD34+ HPC samples isolated from either peripheral blood or bone marrow from 15 healthy donors were shown to express high telomerase activity. However, telomerase activity was undetectable in nine and severely impaired in two CD34+ HPC populations isolated from the peripheral blood from 11 HIV-1-infected patients.

The function of telomerase in HPC is somewhat controversial [16], and there is no consensus on which HPC subpopulation or physiological condition is best suited for enzyme expression [20,21]. In addition, telomerase activity also seems to be present in mature T and B cells [21,31], whereas an ageand proliferation-related loss of telomeric DNA has been observed in haematopoietic stem cells [32].

Nevertheless, our data, which showed dramatically impaired telomerase levels in CD34+ HPC isolated from the peripheral blood of HIV-1-seropositive patients, unequivocally disclosed an additional phenotypic character of CD34+ HPC in HIV-1-infected patients. These findings may have some relevance in the pathogenesis of lesions in the HPC compartment.

We have also shown that CD34+ HPC isolated from healthy subjects show significantly impaired telomerase activity (P < 0.001) when exposed to rgp120 or, to a lesser extent, TGF-β1. These results may help explain our previous findings that purified normal CD34+ HPC show an impaired survival/proliferation capacity after exposure to heat-inactivated HIV-1 or gp120 [4,6]. They are also consistent with the possible role of endogenous TGF-β1 in mediating the inhibitory effect of either HIV-1 or gp120 [9,10] and the demonstration that TGF-β1 is an inhibitor of telomerase activity [14]. Together, these results point to a possible direct intervention of soluble or virus-associated gp120, as the cause of the severely impaired telomerase activity exhibited by HPC isolated from HPC-1-infected patients. In this respect, we have already demonstrated an early loss of circulating HPC during the course of HIV-1 infection [33] and a close correlation between the impaired number of circulating HPC and active HIV-1 replication [34].

The results of our experiments demonstrate severely impaired telomerase expression in uninfected CD34+ HPC isolated from the peripheral blood of HIV-1-infected patients. This finding was irrespective of concurrent antiretroviral therapy; therefore, the possible influence of RT inhibitor therapy on the levels of telomerase activity can be ruled out. At the same time, a long-standing consequence of previous zidovudine treatments seems unlikely. In the Tetrahymena termophila model, zidovudine was one of the least efficient telomerase inhibitors amongst the various nucleoside analogues [35]. In immortalized human cell lines, zidovudine was only effective in significantly reducing telomerase activity when employed directly in the reaction mixture at very high concentrations (50–100 mmol/l) [36], far beyond the levels that can be reached in HIV-1-infected patients, even with a chronic dosing of 250 mg every 4 h (peak and trough concentrations of 0.6 and 0.16 µg/ml plasma, respectively). In vivo zidovudine has a very short half-life and is rapidly metabolized to 3′-azido-3′-deoxy-5′-O-β-D-glucopyranuronosylthymidine, which has no effect as an RT inhibitor. The influence of zidovudine on telomerase function and cell senescence of immortalized mouse fibroblasts is completely reversible after inhibitor removal [37]. Six out of 11 HIV-1-infected patients studied did not receive antiretroviral therapy with RT inhibitors for at least 5 months and the lack of telomerase activity observed in HIV-1-infected patients was irrespective of ongoing antiretroviral therapy. Finally, our results showed that gp120 and, to a lesser extent, TGF-β1 are very efficient inhibitors of telomerase activity (much more efficient, on a molar basis, than zidovudine). All these considerations, in our opinion, suggest that the impaired telomerase activity of CD34+ cells of HIV-1-infected subjects is very likely to be a virus-mediated phenomenon rather than a long-standing consequence of zidovudine therapy.

Therefore, we can conclude that the mechanism underlying the impaired telomerase activity of HPC is likely to involve the interaction of HIV-1 envelope glycoprotein gp120 with the cell membrane, and possibly the upregulation of endogenous TGF-β1 expression. These findings may further explain the severely compromised survival/replication capacities [1,2] and the enhanced propensity to apoptosis [6] of HPC in HIV-1-infected patients, despite the lack of productive/latent HIV-1 infection [1,2].

Whether endogenous TGF-β1 represents an essential intermediate in the gp120-mediated inhibition of telomerase expression remains to be fully elucidated. Further studies are also necessary to establish the relevance of telomerase in the replicative potential of HPC and the role of its inhibition in the pathogenesis of their altered phenotype during HIV-1 infection. These studies, amongst others, may have profound implications in judging the feasibility of experiments aimed at conferring HIV-1 resistance by genetically engineering haematopoietic stem/progenitor cells.

References

1. Zauli G, Davis BR: Role of HIV infection in the hematologic manifestations of HIV seropositive subjects. Crit Rev Oncol Hematol 1993, 15:271–283.

2. Re MC, Furlini G, Zauli G, La Placa M: Human immunodeficiency virus type 1 (HIV-1) and human hematopoietic progenitors. Brief review. Arch Virol 1994, 137:1–23.

3. Zauli G, Re MC, Davis B, et al.: Impaired in vitro growth of purified (CD34+) hematopoietic progenitors in human immunodeficiency virus-1 seropositive thrombocytopenic individuals. Blood 1992, 79:2680–2687.

4. Zauli G, Re MC, Furlini G, Giovannini M, La Placa M: Human immunodeficiency virus type 1 envelope glycoprotein gp120-mediated killing of human hematopoietic progenitors (CD34+ cells). J Gen Virol 1992, 73:417–421.

5. Re MC, Zauli G, Gibellini D, et al.: Uninfected hematopoietic progenitor (CD34+) cells purified from the bone marrow of AIDS patients are committed to apoptotic cell death in culture. AIDS 1993, 7:1049–1055.

6. Zauli G, Vitale M, Re MC, et al.: In vitro exposure to human immunodeficiency virus type 1 induces apoptotic death of the factor-dependent TF-1 hematopoietic cell line. Blood 1994, 83:167–175.

7. Zauli G, Furlini G, Vitale M, et al.: A subset of human CD34+ hematopoietic progenitors express low levels of CD4, the high-affinity receptor for human immunodeficiency virus type 1. Blood 1994, 84:1896–1905.

8. Louache F, Debili N, Marandin A, Coulombel L, Vainchenker W: Expression of CD4 by human hematopoietic progenitors. Blood 1994, 84:3344–3355.

9. Zauli G, Vitale M, Gibellini D, Capitani S: Inhibition of purified CD34+ hematopoietic progenitor cells by human immunodeficiency virus 1 or gp120 mediated by endogenous transforming growth factor β1. J Exp Med 1996, 183:99–108.

10. Gibellini D, Celeghini C, Panaya R, et al.: The engagement of CD4 surface antigen in the HEL haematopoietic cell line up-regulates the transforming growth factor-β1 (TGF-β1) promoter activity. Br J Haematol 1997, 97:571–578.

11. Lardon F, Snoeck HW, Nijs G, et al.: **Transforming growth factor-**β regulates the cell cycle status of interleukin-3 (IL-3) plus IL-1, stem cell factor, or IL-6 stimulated CD34+ human hematopoietic progenitor cells through different kinetic mechanisms depending on the applied stimulus. Exp Hematol 1994, 22:903–909.

12. Li CY, Suardet L, Littel JB: Potential role of WAF1/Cip1/p21 as mediator of TGF-beta cytoinhibitory effect. J Biol Chem 1995, 270:4971–4974.

13. Selvakumaran M, Lin HK, Miyashita T, et al.: Immediate early up-regulation of bax expression by p53 but not TGFβ1: a paradigm for distinct apoptotic pathways. Oncogene 1994, 9:1791–1798.

14. Zhu X, Kumar R, Mandal M, et al.: Cell cycle-dependent modulation of telomerase activity in tumor cells. Proc Natl Acad Sci USA 1996, 93:6091–6095.

15. Blackburn EH: Structure and function of telomeres. Nature 1991, 350:569–573.

16. Greider CW: Telomere length regulation. Annu Rev Biochem 1996, 65:337–365.

17. Harley CB, Kim NW, Prowse KR, et al.: Telomerase, cell immortality, and cancer. Cold Spring Harb Symp Quant Biol 1994, LVIX:307–315.

18. Blasco MA, Lee H-W, Hande MP, et al.: Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 1997, 91:25–34.

19. Morrison SJ, Wandycz AM, Akashi K, Globerson A, Weissman IL: The aging of hematopoietic stem cells. Nature Med 1996, 2:1011–1016.

20. Chiu CP, Dragowska K, Kim NW, et al.: Differential expression of telomerase activity in hematopoietic progenitors from adult human bone marrow. Stem Cells 1996, 14:239–248.

21. Hiyama K, Hirai Y, Kyaizumi S, et al.: Activation of telomerase in human lymphocytes and hematopoietic progenitor cells. J Immunol 1995, 155:3711–3715.

22. Sutherland DR, Keating A, Nayar R, et al.: Sensitive detection and enumeration of CD34+ cells in peripheral and cord blood by flow cytometry. Exp Hematol 1994, 22:1003–1010.

23. Tong J, Hoffman R, Siena S, et al.: Characterization and quantitation of primitive hematopoietic progenitor cells present in peripheral blood autographs. Exp Hematol 1994, 22:1016–1024.

24. de Wynter EA, Coutinho LH, Pei X, et al.: Comparison of purity and enrichment of CD34+ cells from bone marrow, umbilical cord and peripheral blood (primed for apheresis) using five separation systems. Stem Cells 1995, 13:524–532.

25. Kim NW, Piatyszek MA, Prowse KR, et al.: Specific association of human telomerase activity with immortal cells and cancer. Science 1994, 266:2011–2015.

26. Piatyszek MA, Kim NW, Weinrich SL, Hiyama K, Wright WE, Shay JW: Detection of telomerase activity in human cells and tumors by a telomeric repeat amplification protocol (TRAP). Methods Cell Sci 1994, 17:1–15.

27. Centers for Disease Control: 1993 Revised classification system for HIV infection and expanded surveillance case definition for AIDS among adolescents and adults. MMWR 1992, 41:1–19.

28. Effros RB, Allsopp R, Chiu C-P, et al.: Shortened telomeres in the expanded CD28-CD8+ cell subset in HIV disease implicate replicative senescence in HIV pathogenesis. AIDS 1996, 10:F17-F22.

29. Palmer LD, Weng N-P, Levine BL, June CH, Lane CH, Hodes RJ: Telomere length, telomerase activity, and replicative potential in HIV infection: analysis of CD4- and CD8+ T cells from HIV-discordant monozygotic twins. J Exp Med 1997, 185:1381–1386.

30. Pommier JP, Gauthier L, Livartowski J, et al.: Immunosenescence in HIV pathogenesis. Virology 1997, 231:148–154.

31. Broccoli D, Young JW, De Lange T: Telomerase activity in normal and malignant hematopoietic cells. Proc Natl Acad Sci USA 1995, 92:9082–9086.

32. Vaziri H, Dragowska W, Allsopp RC, Thomas TT, Harley CB, Lansdorp PM: Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age. Proc Natl Acad Sci USA 1994, 91:9857–9860.

33. Bagnara GP, Zauli G, Giovannini M, Re MC, Furlini G, La Placa M: Early loss of circulating hematopoietic progenitors in HIV-1-infected subjects. Exp Hematol 1990, 18:426–430.

34. Re MC, Zauli G, Furlini G, et al.: The impaired number of circulating granulocyte/macrophage progenitors (CFU-GM) in human immunodeficiency virus type 1 infected subjects correlates with an active HIV-1 replication. Arch Virol 1993, 129:53–64.

35. Strahl C, Blackburn EH: The effects of nucleoside analogs on telomerase and telomeres in Tetrahymena. Nucleic Acid Res 1994, 22:893–900.

36. Strahl C, Blackburn EH: Effects of reverse transcriptase inhibitors on telomere length and telomerase activity in two immortalized cell lines. Mol Cell Biol 1996, 16:53–65.

37. Yegorov YE, Chernov DN, Akimov SS, Bolsheva NL, Krayeska AA, Zelenin AV: Reverse transcriptase inhibitors suppress telomerase function and induce senescence-like processes in cultured mouse fibroblasts. FEBS Lett 1996, 389:115–118.

Keywords:

Telomerase; haematopoietic progenitor cells; HIV-1 infection; transforming growth factor-β1; gp120

© 1998 Lippincott Williams & Wilkins, Inc.