THROMBOCYTOPENIA, anemia, lymphocytopenia, monocytopenia, and neutropenia and permutations of these abnormalities are found in most patients with acquired immunodeficiency syndrome (AIDS). In virtually all patients with advanced AIDS (group IV), pancytopenia is the rule. More than 90% of such patients are anemic and an equal fraction have either neutropenia or monocytopenia.1 Although immune thrombocytopenia is a common cause of low platelet counts in human immunodeficiency virus (HIV)-infected patients, the majority of other types of cytopenias usually reflect bone marrow (BM) dysfunction. Even so, the causes of these cytopenias are clearly heterogeneous and can, in a large number of cases, be attributed to the widely recognized hematopoietic suppressive effects of certain types of intercurrent infections (eg, cytomegalovirus, parvovirus, hepatitis virus, and mycobacterial infection), or of drugs commonly used in AIDS patients (zidovudine, gancyclovir, trimethoprim/sulfa). The multiple causes of marrow failure in patients with AIDS1-3 account for the mixed histological findings on BM examinations of these patients.1 BM biopsy samples and aspirates are most often nonspecifically abnormal. To be sure, certain findings are more than nonspecific. The discovery of abundant megakaryocytes in the BM of a patient with isolated thrombocytopenia, for example, is highly suggestive of idiopathic thrombocytopenic purpura (ITP). Erythroid hypoplasia suggests either parvovirus infection (a rare cause of anemia in patients with HIV infection4) or infection of the marrow with mycobacterium avium complex.1 Interestingly, frank hypocellularity is the exception and aplasia is rare. Taking these diagnostic complexities into account, it has been impossible, from examination of clinical material alone, to determine whether HIV-1 itself has any direct impact on hematopoietic activity.

We seek in this review not to catalogue the spectrum of hematopoietic defects that occur in patients with AIDS, but to focus specifically on the influence HIV-1 itself imposes on hematopoietic cells. For more than a decade, research teams worldwide have sought to identify a causal role for HIV-1 and its gene products in the hematopoietic defects that develop in patients with AIDS and more clear paradigms have emerged. The most important of these is that the greatest impact of viral infection on growth and differentiation of hematopoietic progenitor cells results from the capacity of the virus to infect and perturb the hematopoietic regulatory function of auxiliary cells, not from its capacity to infect progenitors and stem cells themselves. Here we review the evidence in support of this model. We will focus on two commonly encountered AIDS-related hematopoietic abnormalities in clinical practice, regenerative marrow failure and lymphoid neoplasms. Because BM failure and lymphomas are common consequences of AIDS progression, and limit both survival and quality of life, a clear pathophysiological picture of these disorders and the molecular mechanism(s) by which HIV-1 causes or sets the stage for them, is an essential prerequisite for the development of rational strategies for therapy and prevention.

Recovery from myelosuppression induced by treatment with antivirals and with antilymphoma regimens is consistently more toxic to hematopoietic tissues of AIDS patients than in those of non-AIDS patients. Although many AIDS patients have BM failure before therapy,5-12 a greater number experience inordinate myelosuppressive toxicity after conventional antilymphoma13 or antiviral14chemotherapy unless hematopoietic growth factors are used.14-16 Even when growth factors are used, conventional therapy is extremely toxic. Levine et al17 recently described the results of their study using ABVD (Adriamycin [Pharmacia & Upjohn, Kalamazoo, MI], bleomycin, Velban [Lilly, Indianapolis, IN], and Dacarbazine [Bayer, West Haven, CT]) with granulocyte colony-stimulating factor (G-CSF) support of 21 HIV+ patients with Hodgkin's disease, in which nearly every patient had either severe and prolonged thrombocytopenia and/or neutropenia.17 Theoretically, causes of regenerative failure might reflect dysfunction of the hematopoietic progenitor cell population, dysfunction of cells that control the regenerative response by releasing hematopoietic growth factors, or activation of gene programs that serve to inhibit one or both of these two cell types.18 The weight of evidence today quite clearly suggests that the progenitor populations are intact and largely uninfected, but that auxiliary cells are very consistently infected and broadly dysfunctional.

Infection of Hematopoietic Progenitor Cells Is Infrequent and not Substantially Involved in the Pathogenesis of Marrow Failure

Infection of hematopoietic progenitor cells can be demonstrated in one of a number of ways. One can infect a population of BM cells ex vivo or acquire marrow cells from seropositive patients and then either plate single highly enriched progenitor cells in multi-well plates or plate such cells in semisolid medium. Once wells with colonies are identified, provirus, viral mRNA transcripts, or viral proteins must be detected in all of the cells of the colony and, ideally, infectious virus should be detected in coculture assays. Alternatively, clones arising from plated single progenitor cells plucked from semisolid medium can be used in the same assays. If these steps are not taken, the appearance of infected macrophages in the cultures cannot be attributed to direct progenitor cell infection because they may have simply survived in vitro having been infected in vivo. Enrichment of CD34+ cells before plating in clonal assays does not sidestep this pitfall. Such BM cell populations may also contain microvascular endothelial cells (MVEC) which universally express CD34 and are consistently infected in marrow from seropositive patients.19 Thus, in vitro infection of more differentiated myeloid cells and even T cells20 could result from in vivo infection of contaminating MVEC when CD34+ cells are plated in clonal assays. Using these strict criteria, only 1 of the 36 studies listed in Tables 1 and2convincingly demonstrate that committed progenitor cells can be infected by HIV-1.21 

Table 1.

Reports That Describe the Effects of Ex Vivo HIV-1 Infection on Hematopoietic Cells

Reference Method Progenitor Growth HIV-1 Infection of Progenitors? HIV-1 Infection of Clonal Progeny? Comments
Folks et al173  HIV-1 challenge of CD34+ BM cells. Infectivity evaluated by RT assay and EM.  Not evaluated.  Evidence of HIV-1 infection at 1 mo pi (RT activity). Virus detected by EM at 40-60 d pi.  Not evaluated.  No convincing evidence of progenitor cell infection. HIV-1–infected CD34+ stromal cells or residual T cells/MΘ could infect differentiating BM cells.  
Molina et al27 (see Table 2Unfractionated BM cells challenged by HIV-1 and seeded into colony assays.  Colony growth normal.  Not evaluated.  No HIV-1 DNA detected (gag PCR).  No evidence of progenitor cell infection. 
Steinberg et al174  NATD BM cells and CD34+ BM cells challenged with HIV-1.  ↓ CFU-GM ↓ BFU-E  HIV-1 DNA detected (gag PCR). Signal much weaker in NATD cells.  No HIV-1 DNA detected (gag PCR).  No convincing evidence of progenitor cell infection. Contaminating T cells/MΘ may yield PCR signal. Anti-gp 160 abrogates inhibition of colony growth.  
Kojouharoff et al175 (see Table 2)  Coculture of CD34+ BM cells with HIV-1-infected M and seeding into colony assays.  Colony growth normal.  Not evaluated.  HIV-1 mRNA transcripts in “40% of CFU-GEMM and CFU-GM (in situ hybridization).  No convincing evidence of progenitor cell infection. MΘ-to-MΘ infection could not be excluded.  
Zauli et al176  CD34+ cells from PB infected and seeded into colony assays.  ↓ CFU-GM ↓ BFU-E ↓ CFU-Mk  Not evaluated.  No HIV-1 DNA in pooled colonies (gag PCR).  No evidence of progenitor cell infection.  
Zauli et al177  CD34+ BM cells exposed to recombinant gp160, gp120, gp41, & p24.  ↓ CFU-GM & ↓ CD34+ cells via gp160 & gp120. Not evaluated.  Not evaluated.  Suggests a mechanism for HIV-1-induced cytopenia without direct infection.  
Calenda et al178  Challenge of LTBMC with HIV-1 strains LAV and NDK.  ↓↑ CFU-E ↓↑ BFU-E  Virus recovered from LTBMC by cocultured with PBMC.  Not evaluated.  No convincing evidence of progenitor cell infection. Variable results for colony growth (↓↑) depending on virus strain, moi, and time. 
Steinberg et al8  Human stromal cell line (BS-1) infected with HIV-1 and cocultured with murine BM progenitors.  ↓ BFU-E ↓ CFU-GM  Not evaluated. Murine lineage cells.  Not evaluated. Murine lineage cells. Suppression of murine colony growth attributed to HIV-1 infection of stromal cells.  
Cen et al179  Challenge of LTBMC and CD34+ BM cells with HIV-1 strains LAV, NL4-3 and ICR 3.  ↓ CFU-GM  No HIV-1 DNA detected (gag/env PCR).  Not evaluated.  No evidence of progenitor cell infection. Treatment of inoculum with anti-gp120 abrogates inhibition of colony growth. Infection of cellular elements in LTBMC with T cell-tropic HIV-1. 
Zauli et al33  CD34+ cell line (TF-1) exposed to HIV-1 or gp120 plus anti-gp120 and anti-CD4 antibodies. Not evaluated.  No viral DNA (gag/env PCR) or protein (p24 ELISA).  Not evaluated.  No evidence of progenitor cell infection. Treatment with gp120 & antibodies induced apoptosis; suggests mechanism for cytopenia.  
Maciejewski et al167  Total BM cells, MΘ-depleted BM cells and CD34+ BM cells challenged with HIV-1 or heat-inactivated HIV-1.  ↓ CFU-GM ↓ BFU-E in total BM cultures only.  Not evaluated.  Not evaluated.  Depletion experiments implicate MΘ. Heat-inactivated HIV-1 inhibits colony growth. Inhibition partially blocked by anti-gp120 or anti-TNFα. 
Schwartz et al180  BM stroma challenged with HIV-1 strain ADA and overlaid with CD34+ cells.  ↓ CFU-GM ↓ BFU-E  Not evaluated.  Not evaluated.  Factors from HIV-1-infected stroma supress colony growth. Neutralization of IFNα or IL-4 restores growth. 
Marandin et al61  Challenge of LTBMC with monocyte-tropic HIV-1.  Colony growth normal.  Not evaluated. Not evaluated.  MΘ were only infected stromal cells detected. Cytokine profiles unaffected.  
Chelucci et al21  CD34+/Lin PB cells challenged with HIV-1 and seeded into colony assays.  Colony growth normal.  Not evaluated.  HIV-1 DNA (PCR), RNA (RT-PCR) or protein (p24 ELISA) detected.  CFU-GM and BFU-E from PB infected in vitro. CD34+/CD4+ cells (5-20% of total) were presumed targets.  
Zauli et al172  CD34+ BM cells challenged with HIV-1 or heat-inactivated HIV-1.  ↓ CFU-GM  No HIV-1 DNA detected (gag PCR).  Not evaluated.  No evidence of progenitor cell infection. gp120 induction of TGFb inhibits colony growth. 
Kaushal et al181  CD34+ BM cells (K6.1 MAB selected) challenged with HIV-1. After 24 h reselected for CD34 with ICH3 MAB.  Not evaluated.  No HIV-1 RNA detected (RT-PCR for unspliced and spliced tat-rev-nef transcripts).  Not evaluated. No evidence of progenitor cell infection. Two-step purification enhances purity of CD34 population. 
Reference Method Progenitor Growth HIV-1 Infection of Progenitors? HIV-1 Infection of Clonal Progeny? Comments
Folks et al173  HIV-1 challenge of CD34+ BM cells. Infectivity evaluated by RT assay and EM.  Not evaluated.  Evidence of HIV-1 infection at 1 mo pi (RT activity). Virus detected by EM at 40-60 d pi.  Not evaluated.  No convincing evidence of progenitor cell infection. HIV-1–infected CD34+ stromal cells or residual T cells/MΘ could infect differentiating BM cells.  
Molina et al27 (see Table 2Unfractionated BM cells challenged by HIV-1 and seeded into colony assays.  Colony growth normal.  Not evaluated.  No HIV-1 DNA detected (gag PCR).  No evidence of progenitor cell infection. 
Steinberg et al174  NATD BM cells and CD34+ BM cells challenged with HIV-1.  ↓ CFU-GM ↓ BFU-E  HIV-1 DNA detected (gag PCR). Signal much weaker in NATD cells.  No HIV-1 DNA detected (gag PCR).  No convincing evidence of progenitor cell infection. Contaminating T cells/MΘ may yield PCR signal. Anti-gp 160 abrogates inhibition of colony growth.  
Kojouharoff et al175 (see Table 2)  Coculture of CD34+ BM cells with HIV-1-infected M and seeding into colony assays.  Colony growth normal.  Not evaluated.  HIV-1 mRNA transcripts in “40% of CFU-GEMM and CFU-GM (in situ hybridization).  No convincing evidence of progenitor cell infection. MΘ-to-MΘ infection could not be excluded.  
Zauli et al176  CD34+ cells from PB infected and seeded into colony assays.  ↓ CFU-GM ↓ BFU-E ↓ CFU-Mk  Not evaluated.  No HIV-1 DNA in pooled colonies (gag PCR).  No evidence of progenitor cell infection.  
Zauli et al177  CD34+ BM cells exposed to recombinant gp160, gp120, gp41, & p24.  ↓ CFU-GM & ↓ CD34+ cells via gp160 & gp120. Not evaluated.  Not evaluated.  Suggests a mechanism for HIV-1-induced cytopenia without direct infection.  
Calenda et al178  Challenge of LTBMC with HIV-1 strains LAV and NDK.  ↓↑ CFU-E ↓↑ BFU-E  Virus recovered from LTBMC by cocultured with PBMC.  Not evaluated.  No convincing evidence of progenitor cell infection. Variable results for colony growth (↓↑) depending on virus strain, moi, and time. 
Steinberg et al8  Human stromal cell line (BS-1) infected with HIV-1 and cocultured with murine BM progenitors.  ↓ BFU-E ↓ CFU-GM  Not evaluated. Murine lineage cells.  Not evaluated. Murine lineage cells. Suppression of murine colony growth attributed to HIV-1 infection of stromal cells.  
Cen et al179  Challenge of LTBMC and CD34+ BM cells with HIV-1 strains LAV, NL4-3 and ICR 3.  ↓ CFU-GM  No HIV-1 DNA detected (gag/env PCR).  Not evaluated.  No evidence of progenitor cell infection. Treatment of inoculum with anti-gp120 abrogates inhibition of colony growth. Infection of cellular elements in LTBMC with T cell-tropic HIV-1. 
Zauli et al33  CD34+ cell line (TF-1) exposed to HIV-1 or gp120 plus anti-gp120 and anti-CD4 antibodies. Not evaluated.  No viral DNA (gag/env PCR) or protein (p24 ELISA).  Not evaluated.  No evidence of progenitor cell infection. Treatment with gp120 & antibodies induced apoptosis; suggests mechanism for cytopenia.  
Maciejewski et al167  Total BM cells, MΘ-depleted BM cells and CD34+ BM cells challenged with HIV-1 or heat-inactivated HIV-1.  ↓ CFU-GM ↓ BFU-E in total BM cultures only.  Not evaluated.  Not evaluated.  Depletion experiments implicate MΘ. Heat-inactivated HIV-1 inhibits colony growth. Inhibition partially blocked by anti-gp120 or anti-TNFα. 
Schwartz et al180  BM stroma challenged with HIV-1 strain ADA and overlaid with CD34+ cells.  ↓ CFU-GM ↓ BFU-E  Not evaluated.  Not evaluated.  Factors from HIV-1-infected stroma supress colony growth. Neutralization of IFNα or IL-4 restores growth. 
Marandin et al61  Challenge of LTBMC with monocyte-tropic HIV-1.  Colony growth normal.  Not evaluated. Not evaluated.  MΘ were only infected stromal cells detected. Cytokine profiles unaffected.  
Chelucci et al21  CD34+/Lin PB cells challenged with HIV-1 and seeded into colony assays.  Colony growth normal.  Not evaluated.  HIV-1 DNA (PCR), RNA (RT-PCR) or protein (p24 ELISA) detected.  CFU-GM and BFU-E from PB infected in vitro. CD34+/CD4+ cells (5-20% of total) were presumed targets.  
Zauli et al172  CD34+ BM cells challenged with HIV-1 or heat-inactivated HIV-1.  ↓ CFU-GM  No HIV-1 DNA detected (gag PCR).  Not evaluated.  No evidence of progenitor cell infection. gp120 induction of TGFb inhibits colony growth. 
Kaushal et al181  CD34+ BM cells (K6.1 MAB selected) challenged with HIV-1. After 24 h reselected for CD34 with ICH3 MAB.  Not evaluated.  No HIV-1 RNA detected (RT-PCR for unspliced and spliced tat-rev-nef transcripts).  Not evaluated. No evidence of progenitor cell infection. Two-step purification enhances purity of CD34 population. 

Abbreviations: NATD, nonadherent T-cell depleted; MΘ, macrophage; EM, electron microscopy; PB, peripheral blood; pi, postinfection; LTBMC, long-term bone marrow cultures.

Table 2.

Reports That Describe Hematopoietic Function of Cells Obtained From HIV-1–Seropositive Patients

Reference Method Progenitor Growth HIV-1 Infection of Progenitors? HIV-1 Infection of Clonal Progeny? Comments
Stella et al40  BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-GEM ↓ BFU-E ↓ CFU-Mk  Not evaluated.  Not evaluated.  Depletion of T cells enhanced colony growth. Suggest a T-cell imbalance may contribute to impaired hematopoiesis. 
Donahue et al182  BM cells from HIV-1+patients seeded into colony assays in the presence/absence of anti–HIV-1 antisera.  ↓ CFU-GM & ↓ BFU-E only in presence of antisera.  Not evaluated.  HIV-1 in pooled colonies from 1/4 patients (RT activity in culture supernatants). No convincing evidence of progenitor cell infection. More differentiated cells in culture may be virus source. Anti-gp120 antibodies may be suppressive elements in patient sera. 
Lunardi-Iskandar et al11  BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  Not evaluated.  Not evaluated.  Depletion of adherent cells or T cells or addition of cytokines (GM-CSF, IL-3, Epo) did not restore colony growth). 
Molina et al27 (see Table 1)  BM cells from HIV-1+ patients seeded into colony assays.  Colony growth normal.  Not evaluated.  No HIV-1 DNA detected (gag PCR). No evidence of progenitor cell infection. Propose cytopenias due to alteration in BM microenvironment.  
Von Laer et al183  Culture of purified CD34+ BM cells from HIV-1+ patients.  Not evaluated.  HIV-1 DNA detected in 1/14 samples (gag PCR).  Not evaluated.  No convincing evidence of progenitor cell infection. Single positive attributed to CD4+ T-cell contamination.  
Ganser et al184  BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-GEM ↓ BFU-E ↓ CFU-Mk Not evaluated.  No HIV-1 detected (immunohistochemistry and in situ hybridization).  No evidence of progenitor cell infection. 
Davis et al28  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-E  HIV-1 DNA detected in 1/11 samples (gag/env PCR).  No HIV-1 DNA detected (gag/env PCR).  No convincing evidence of progenitor cell infection. Single positive attributed to CD4+ T-cell or MΘ contamination. 
Kojouharoff et al175 (see Table 2)  BM cells from HIV-1+ patients seeded into colony assays.  Colony growth normal.  Not evaluated.  HIV-1 mRNA detected in CFU-GEM & CFU-GM in 2/6 patients (in situ hybridization)  No convincing evidence of progenitor cell infection. HIV-1+ cells may reflect infection of a more differentiated phenotype.  
Zauli et al25  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-E ↓ CFU-Mk No HIV-1 DNA (gag PCR) or protein (p24 ELISA) detected.  Not evaluated.  No evidence of progenitor cell infection. HIV-1 detected in unfractionated bone marrow samples. A role for HIV-1 in the BM microenvironment proposed.  
Zauli et al185  Purified CD34+ BM cells from HIV-1+patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-Mk  HIV-1 DNA detected in 2/17 samples (gag/env PCR). No p24 in culture supernatants.  Not evaluated.  No convincing evidence of progenitor cell infection. Two positives attributed to CD4+ T-cell or MΘ contamination. 9 CFU-Mk in all samples implies an indirect mechanism.  
Stanley et al150  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  HIV-1 DNA (gag/env PCR) & virus (co-culture) detected in 17/52 & 8/29 Zairian and 3/22 & 1/2 American samples.  Not evaluated.  HIV-1 detected only in subset of patients with advanced disease. 9 CFU-GM & 9 BFU-E in all samples implies an indirect mechanism. CD34+ accessory cells are potential source of HIV-1.  
Louache et al186  BM cells from 27 HIV-1+ patients seeded into colony assays. ↓ CFU-GM ↓ BFU-E ↓ CFU-Mk  Not evaluated.  No HIV-1 DNA detected (gag PCR).  No evidence of progenitor cell infection. Anti-sense nef or tat increased clonal growth.  
Kaczmarski et al187  BM cells from HIV-1+ patients studied in LTBMC and colony assays. Normal CFU-GEMM and CFU-GM.  Not evaluated.  HIV-1 DNA detected in pooled CFU-GEMM, CFU-GM & CFU-LTC (gag/env/tat PCR).  No convincing evidence of progenitor cell infection. No T-cell depletion so infection of differentiating cells could not be ruled out. 
De Luca et al29  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  No HIV-1 DNA detected (gag PCR).  Not evaluated.  No evidence of progenitor cell infection.  
Hillyer et al7  Purified CD34+ cells from SIV-infected macaques seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  No SIV DNA detected (gag PCR).  SIV DNA detected in occasional pooled CFU-GM colonies.  No convincing evidence of progenitor cell infection. Rare positive signal attributed to CD4+ T-cell or MΘ contamination.  
Neal et al34  Purified CD34+ BM cells from HIV-1+ patients studied in LTBMC and colony assays.  Normal CFU-E, BFU-E, CFU-GM & cobblestone formation in LTBMC.  HIV-1 DNA detected in 2/10 patients (gag/pol PCR).  All pooled CFU-GM were HIV-1 DNA negative (gag/pol PCR). 1/5 pooled CFU-E/BFU-E were weakly HIV-1+ (<2 copies/75,000 cells).  No convincing evidence of progenitor cell infection. Low proviral DNA copy number in pooled colonies (2-5 copies/250,000 cells) suggests (CD4+ T-cell or MΘ contamination. 
Dallalio et al188  Purified CD34+ BM cells from 5 HIV-1+ patients seeded into CFU-E assays Normal CFU-E.  Not evaluated.  Not evaluated.  CFU-E were not more sensitive to cytokine-mediated inhibition of colony formation.  
Slobod et al31  Mobilization of CD34+ BM cells from HIV-1+ patients. Nested PCR for evidence of HIV-1 infection.  Not evaluated.  HIV-1 DNA in 2/7 patients but in only 1/3 replicate samples (nested env PCR detecting 1 DNA copy/25,000 cells).  Not evaluated  No convincing evidence of progenitor cell infection. CD34+ cells at 92-99% purity so variable positives attributed to CD4+T-cell contamination.  
Marandin et al189  Primitive BM cells from HIV-1+ patients evaluated by flow cytometry. Fractionated CD34+ cells studied in LTBMC and colony assays.  ↓ CFU-GM ↓ BFU-E ↓ LTC-IC  No HIV-1 DNA detected (gag PCR). Included separate analysis of CD34+/CD4+, CD34+/CD38+ & CD34+/CD38 populations.  No HIV-1 DNA detected (gag PCR).  No significant change in percentage of CD34+ cells in HIV-1+ patients but proportions of CD34+/CD38 & CD34+/Thy 1+ cells decreased. Suggests loss of more primitive phenotype but not via direct infection.  
Kearns et al151  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM HIV-1 DNA detected in rare samples. Semi-quantitative PCR estimates 1/100,000 cells infected.  Not evaluated.  No evidence of significant progenitor cell infection. Propose dysregulated hematopoiesis due to alterations in the BM microenvironment. 
Bahner et al52  Ex vivo infection of human marrow stromal cells with HIV-1 and stomal cells genetically modified to resist HIV-1 infection. Stroma overlayered with bone marrow cells. ↓ CFU-GM (obtained after 3 wk on stroma)  Not evaluated. Not evaluated.  Stroma is infectable. Reduced capacity of HIV-1-infected stroma to support hematopoietic progenitor cell growth. 
Reference Method Progenitor Growth HIV-1 Infection of Progenitors? HIV-1 Infection of Clonal Progeny? Comments
Stella et al40  BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-GEM ↓ BFU-E ↓ CFU-Mk  Not evaluated.  Not evaluated.  Depletion of T cells enhanced colony growth. Suggest a T-cell imbalance may contribute to impaired hematopoiesis. 
Donahue et al182  BM cells from HIV-1+patients seeded into colony assays in the presence/absence of anti–HIV-1 antisera.  ↓ CFU-GM & ↓ BFU-E only in presence of antisera.  Not evaluated.  HIV-1 in pooled colonies from 1/4 patients (RT activity in culture supernatants). No convincing evidence of progenitor cell infection. More differentiated cells in culture may be virus source. Anti-gp120 antibodies may be suppressive elements in patient sera. 
Lunardi-Iskandar et al11  BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  Not evaluated.  Not evaluated.  Depletion of adherent cells or T cells or addition of cytokines (GM-CSF, IL-3, Epo) did not restore colony growth). 
Molina et al27 (see Table 1)  BM cells from HIV-1+ patients seeded into colony assays.  Colony growth normal.  Not evaluated.  No HIV-1 DNA detected (gag PCR). No evidence of progenitor cell infection. Propose cytopenias due to alteration in BM microenvironment.  
Von Laer et al183  Culture of purified CD34+ BM cells from HIV-1+ patients.  Not evaluated.  HIV-1 DNA detected in 1/14 samples (gag PCR).  Not evaluated.  No convincing evidence of progenitor cell infection. Single positive attributed to CD4+ T-cell contamination.  
Ganser et al184  BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-GEM ↓ BFU-E ↓ CFU-Mk Not evaluated.  No HIV-1 detected (immunohistochemistry and in situ hybridization).  No evidence of progenitor cell infection. 
Davis et al28  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-E  HIV-1 DNA detected in 1/11 samples (gag/env PCR).  No HIV-1 DNA detected (gag/env PCR).  No convincing evidence of progenitor cell infection. Single positive attributed to CD4+ T-cell or MΘ contamination. 
Kojouharoff et al175 (see Table 2)  BM cells from HIV-1+ patients seeded into colony assays.  Colony growth normal.  Not evaluated.  HIV-1 mRNA detected in CFU-GEM & CFU-GM in 2/6 patients (in situ hybridization)  No convincing evidence of progenitor cell infection. HIV-1+ cells may reflect infection of a more differentiated phenotype.  
Zauli et al25  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-E ↓ CFU-Mk No HIV-1 DNA (gag PCR) or protein (p24 ELISA) detected.  Not evaluated.  No evidence of progenitor cell infection. HIV-1 detected in unfractionated bone marrow samples. A role for HIV-1 in the BM microenvironment proposed.  
Zauli et al185  Purified CD34+ BM cells from HIV-1+patients seeded into colony assays.  ↓ CFU-GM ↓ CFU-Mk  HIV-1 DNA detected in 2/17 samples (gag/env PCR). No p24 in culture supernatants.  Not evaluated.  No convincing evidence of progenitor cell infection. Two positives attributed to CD4+ T-cell or MΘ contamination. 9 CFU-Mk in all samples implies an indirect mechanism.  
Stanley et al150  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  HIV-1 DNA (gag/env PCR) & virus (co-culture) detected in 17/52 & 8/29 Zairian and 3/22 & 1/2 American samples.  Not evaluated.  HIV-1 detected only in subset of patients with advanced disease. 9 CFU-GM & 9 BFU-E in all samples implies an indirect mechanism. CD34+ accessory cells are potential source of HIV-1.  
Louache et al186  BM cells from 27 HIV-1+ patients seeded into colony assays. ↓ CFU-GM ↓ BFU-E ↓ CFU-Mk  Not evaluated.  No HIV-1 DNA detected (gag PCR).  No evidence of progenitor cell infection. Anti-sense nef or tat increased clonal growth.  
Kaczmarski et al187  BM cells from HIV-1+ patients studied in LTBMC and colony assays. Normal CFU-GEMM and CFU-GM.  Not evaluated.  HIV-1 DNA detected in pooled CFU-GEMM, CFU-GM & CFU-LTC (gag/env/tat PCR).  No convincing evidence of progenitor cell infection. No T-cell depletion so infection of differentiating cells could not be ruled out. 
De Luca et al29  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  No HIV-1 DNA detected (gag PCR).  Not evaluated.  No evidence of progenitor cell infection.  
Hillyer et al7  Purified CD34+ cells from SIV-infected macaques seeded into colony assays.  ↓ CFU-GM ↓ BFU-E  No SIV DNA detected (gag PCR).  SIV DNA detected in occasional pooled CFU-GM colonies.  No convincing evidence of progenitor cell infection. Rare positive signal attributed to CD4+ T-cell or MΘ contamination.  
Neal et al34  Purified CD34+ BM cells from HIV-1+ patients studied in LTBMC and colony assays.  Normal CFU-E, BFU-E, CFU-GM & cobblestone formation in LTBMC.  HIV-1 DNA detected in 2/10 patients (gag/pol PCR).  All pooled CFU-GM were HIV-1 DNA negative (gag/pol PCR). 1/5 pooled CFU-E/BFU-E were weakly HIV-1+ (<2 copies/75,000 cells).  No convincing evidence of progenitor cell infection. Low proviral DNA copy number in pooled colonies (2-5 copies/250,000 cells) suggests (CD4+ T-cell or MΘ contamination. 
Dallalio et al188  Purified CD34+ BM cells from 5 HIV-1+ patients seeded into CFU-E assays Normal CFU-E.  Not evaluated.  Not evaluated.  CFU-E were not more sensitive to cytokine-mediated inhibition of colony formation.  
Slobod et al31  Mobilization of CD34+ BM cells from HIV-1+ patients. Nested PCR for evidence of HIV-1 infection.  Not evaluated.  HIV-1 DNA in 2/7 patients but in only 1/3 replicate samples (nested env PCR detecting 1 DNA copy/25,000 cells).  Not evaluated  No convincing evidence of progenitor cell infection. CD34+ cells at 92-99% purity so variable positives attributed to CD4+T-cell contamination.  
Marandin et al189  Primitive BM cells from HIV-1+ patients evaluated by flow cytometry. Fractionated CD34+ cells studied in LTBMC and colony assays.  ↓ CFU-GM ↓ BFU-E ↓ LTC-IC  No HIV-1 DNA detected (gag PCR). Included separate analysis of CD34+/CD4+, CD34+/CD38+ & CD34+/CD38 populations.  No HIV-1 DNA detected (gag PCR).  No significant change in percentage of CD34+ cells in HIV-1+ patients but proportions of CD34+/CD38 & CD34+/Thy 1+ cells decreased. Suggests loss of more primitive phenotype but not via direct infection.  
Kearns et al151  Purified CD34+ BM cells from HIV-1+ patients seeded into colony assays.  ↓ CFU-GM HIV-1 DNA detected in rare samples. Semi-quantitative PCR estimates 1/100,000 cells infected.  Not evaluated.  No evidence of significant progenitor cell infection. Propose dysregulated hematopoiesis due to alterations in the BM microenvironment. 
Bahner et al52  Ex vivo infection of human marrow stromal cells with HIV-1 and stomal cells genetically modified to resist HIV-1 infection. Stroma overlayered with bone marrow cells. ↓ CFU-GM (obtained after 3 wk on stroma)  Not evaluated. Not evaluated.  Stroma is infectable. Reduced capacity of HIV-1-infected stroma to support hematopoietic progenitor cell growth. 

Abbreviations: NATD, nonadherent T-cell depleted; MΘ, macrophage.

It is important to emphasize that cells expressing CD34 on their surface are not all progenitor cells. Most committed progenitors express CD34 but only 10% to 20% of CD34+ cells from marrow will form colonies. Consequently, when studies are performed on CD34+ cells rather than their clonal progeny, only negative results are informative, vis-à-vis progenitor cell infection. In a frequently cited report, Folks et al173 reported that more than 1 month after ex vivo infection of CD34+ BM cells, monocyte progeny were productively infected by HIV-1. Although the report claimed to have documented infection of purified “progenitor” cells, and while the monocytes that remained in the cultures were likely derived from more primitive precursors, no direct test of progenitor cell infectivity was performed. In fact, in the past few years, most groups have reported that CD34+ cells and/or colony-forming units from seropositive patients are uninfected23-29 (see Table 2), results that nicely match studies performed on rhesus macaques infected with SIV smm9.7 

Theoretically, the failure to discover infected progeny in clonal analyses might result from the induction of apoptosis in infected cells, but the weight of evidence is against this possibility. For example, although in the steady-state circulating committed progenitor cells (colony-forming unit megakaryocyte [CFU-Mk], CFU-granulocyte macrophage [CFU-GM], and burst-forming unit-erythroid [BFU-E]) are reduced in the blood of HIV-1 seropositive patients,30 progenitor reserves are sufficient to be released normally from hematopoietic niches after stimulation. Specifically, HIV-1–infected patients, even patients with advanced disease,31 respond appropriately to exogenous G-CSF by mobilizing progenitor cells of multiple lineages.31,32 More direct in vitro evidence exists against the notion of apoptosis in infected cells. For example, growth of a CD34+ progenitor cell line suppressed by exposure to HIV-1 can be rescued by interleukin-3 (IL-3) and GM-CSF.33 Finally, very direct evidence that progenitor cell apoptosis does not result from HIV-1 infection of progenitors can be found in the only published work that reports stringent progeny analysis from cultures of single cells, in which the potential impact of CD34+ auxiliary cells was minimized. Specifically, Chelucci et al21 purified CD34+/Lin− cells from peripheral blood of healthy volunteers, cultured the cells at densities of 104/mL in the presence of saturating quantities of Steel factor, erythropoietin (Epo), GM-CSF, and IL-3, pelleted the cells, and resuspended them in 50 mL in the presence of HIV-1, HIVIIIB, or NL4-3 strains for 2 hours. The cells were then diluted to 200 mL, cultured overnight, and were then washed and plated in methylcellulose medium at a density of one cell per plate. Colonies were plucked for p24 analysis (enzyme-linked immunosorbent assay [ELISA]) and for HIV-1 tat mRNA reverse transcriptase-polymerase chain reaction (RT-PCR). Twelve percent of CFU-GM colonies were p24 antigen+ and 23% were positive for tat mRNA. Seventeen percent of BFU-E colonies were tat mRNA+ but only 1 of 37 clones was positive for p24. No CFU-GEMM progeny were positive for virus. Appropriate control vectors, including heat-inactivated virus, were used. Consequently, under these clearly defined ex vivo conditions, progenitor cells are infectable, but, more importantly for further consideration of the question of marrow failure, HIV-1 infection ex vivo does not impair the capacity of either CFU-GM or BFU-E to undergo clonal replication or differentiation because differentiated colonial progeny contain the provirus.

Notwithstanding the potential for ex vivo infection, unambiguous evidence for the existence of HIV-1–infected progenitor cells from patients has not yet been described. Based on the above observations and a review of the reports outlined in Tables 1 and 2, it is safe to conclude that: (1) a low fraction of progenitor cells are infectable ex vivo by HIV-1 under some conditions, (2) the growth of the few cells infected by HIV-1 may not be impaired as a result of the infection, and (3) in vivo infection of progenitor cells occurs rarely if ever. There is, on the other hand, clear-cut evidence that viral infection of auxiliary cells and viral gene products themselves can indirectly influence survival and growth of hematopoietic progenitors.

Infected Auxiliary Cells and Regenerative Failure

T cells.

The release of hematopoietic growth factors by T cells is variably influenced by HIV-1 infection. Although IL-6 production is increased,34 production of IL-2 is reduced by HIV-135 possibly via the effects of HIV-1 nef,36 which is known to bind protein kinase Cζ (PKCζ),37 an essential signaling factor in T-cell activation.38,39 It was discovered early on that marrow T cells from HIV-1 patients suppress clonal growth of progenitors in vitro40 but this will be discussed below.

Monocyte/macrophages.

HIV-1 tat and gp120 induce cytokine expression in monocytes, but whether HIV-1 infection of monocytes achieves the same level of induction is arguable. Molina et al41 report that peripheral blood mononuclear cells (PBMC) infected with myriad strains of HIV-1 do not express IL-1, IL-6, or tumor necrosis factor-α (TNF-α). IL-12 release is impaired in infected cells,42and reduced production of IL-1 has been reported.43 Taking into account the important capacity of IL-1 to induce multilineage hematopoietic growth factors,44 and of IL-12 to influence interferon-γ (IFN-γ) expression,45,46 substantial effects on the kinetics of myeloid regeneration could result. The most compelling evidence supporting a role for macrophage dysfunction in regenerative failure is work reported recently on the reduced capacity of monocyte/macrophages to support on-demand myelopoieses after HIV-1 infection.47 Specifically, Esser et al47infected PB-derived monocytes on teflon membranes with HIV-1 (HIV-1D117IIII) and quantified both constitutive and endotoxin-induced secretion of a variety of cytokines. They found that while secretion of TNF-α, IL-1β, IL-6, and IL-8 was stimulated, expression of M, G-, and GM-CSF was inhibited.47 

Microvascular endothelial cells (MVEC).

MVEC from a variety of organs including the brain,48,49liver,50 kidney,51 and BM3,19 are permissive for HIV-1 infection. Of greater relevance to the issue of regenerative marrow failure, BM MVEC cells are always found to be infected by HIV-1 in seropositive patients regardless of the stage of their disease.19 Moreover, while constitutive production of hematopoietic growth factors by such cells (either alone or admixed with other BM stromal cell types) is normal, IL-1 induced production of G-CSF and IL-6 is significantly reduced.19 This suggests that HIV-1 infection of BM MVEC reduces the capacity of hematopoietic stroma to respond to regulatory signals that normally augment blood cell production during periods of increased demand. The dysfunction in marrow stroma does not seem to derive simply from direct interdiction of IL-1 responses in MVEC because pure populations of MVEC, at least from brain, do not respond abnormally to IL-1 after infection (Moses et al, unpublished, April 1997). Therefore, it is likely that the MVEC infection induces release of factors that influence the behavior of other IL-1–responsive cells in the stroma, most of which are not infected (Fig 1). The mechanisms by which this occurs are currently under investigation in our laboratories. The recent report of hematopoietic support dysfunction of HIV-1–infected mixed BM stromal cell cultures containing only 2% MVEC52 supports the notion that collaboration between infected and uninfected cells results in the failure of the stroma to support myeloid hematopoiesis.

Fig. 1.

HIV-1 inhibits replication of hematopoietic progenitor cells (HPC) by reducing production of hematopoietic growth factors. HIV-1 infection of mononuclear phagocytes (M) reduces LPS-induced GM-CSF, M-CSF, and IL-1.47 HIV-1 induces expression of IL-10 in mononuclear leukocytes152,153 and IL-10 inhibits LPS induced release of IL-6, IL-1, GM-CSF, and G-CSF.154,155 Infected MVEC inhibit the responsiveness of BM stromal cells to IL-1 such that IL-6 and G-CSF release is inhibited.19 The mechanism of this effect is unknown and may involve release of a repressor factor (marked in the figure as “?”). Finally, infected macrophages release less IL-143,156 and, because IL-1 controls expression of multiple hematopoietic growth factors in auxiliary cells, including endothelial (MVEC) and other stromal cells (S), the responsiveness of HPC to a given set of environmental cues is functionally reduced.

Fig. 1.

HIV-1 inhibits replication of hematopoietic progenitor cells (HPC) by reducing production of hematopoietic growth factors. HIV-1 infection of mononuclear phagocytes (M) reduces LPS-induced GM-CSF, M-CSF, and IL-1.47 HIV-1 induces expression of IL-10 in mononuclear leukocytes152,153 and IL-10 inhibits LPS induced release of IL-6, IL-1, GM-CSF, and G-CSF.154,155 Infected MVEC inhibit the responsiveness of BM stromal cells to IL-1 such that IL-6 and G-CSF release is inhibited.19 The mechanism of this effect is unknown and may involve release of a repressor factor (marked in the figure as “?”). Finally, infected macrophages release less IL-143,156 and, because IL-1 controls expression of multiple hematopoietic growth factors in auxiliary cells, including endothelial (MVEC) and other stromal cells (S), the responsiveness of HPC to a given set of environmental cues is functionally reduced.

Close modal

Uninfected (bystander) Auxiliary Cells and Regenerative Marrow Failure

T cells.

The clonal suppressive effect of CD8+ hematopoietic inhibitory T cells (HIT cells) has been recognized for decades.53,54 Such cells have been discovered in HIV-1–infected patients.55 Specifically, clonal growth of CFU-GEMM, BFU-E, and CFU-GM from marrows of HIV-1+ patients significantly increased after depletion of CD8+, γδ+, and d-TCS-1+ T cells. No such effect was seen in cultures of normal marrow samples. Further experiments showed that direct cellular contact between effector and hematopoietic progenitor cells was essential and that IFN-γ and TNF-α were key factors mediating the suppressive effect of the d-TCS-1+cells in HIV+ patients.55 In situ hybridization could not detect mRNA from HIV in d-TCS-1+ cells. Therefore, this subset of T cells is an important inhibitor of hematopoietic progenitor cell growth, but they are not infected by HIV-1 (Fig 2).

Fig. 2.

HIV-1 inhibits replication of HPC by inducing release of multiple mitotic inhibitory factors and expression of factors known to induce apoptosis. HIV-1–infected CD4+ T cells and HIV-1 tat released from infected cells induce release of IFN-γ96,157 and TNF-α158 from uninfected CD8+ T cells. HIV-1 and HIV-1 tat induce expression of IL-4 (which inhibits release of hematopoietic growth factors by other cells) and IL-4 receptors (IL-4R) thereby enhancing the “blunting” effect in lymphocytes159,160 and possibly stromal cells. IL-4 also induces stromal cells to release TGF-β161 and chemokine inhibitors.162 HIV-1 induction of fas ligand (FasL) in mononuclear phagocytes (M) and CD4+ cells results in apoptosis of uninfected CD4+ cells,163,164 but may also result in apoptosis of HPC which are known to express Fas under a variety of conditions. 57-59,165,166 Infection of macrophages also induces release of TNF-α,167,168TGF-β,169,170 and MIP1α.171 CD34+cells exposed to HIV-1 or HIV-1 gp120 release TGF-β,172 but while this has been called an “autocrine” response, it is not known whether the TGF-producing cells are actually progenitor cells, and is not, therefore, known to be a truly autocrine mechanism of HPC inhibition.

Fig. 2.

HIV-1 inhibits replication of HPC by inducing release of multiple mitotic inhibitory factors and expression of factors known to induce apoptosis. HIV-1–infected CD4+ T cells and HIV-1 tat released from infected cells induce release of IFN-γ96,157 and TNF-α158 from uninfected CD8+ T cells. HIV-1 and HIV-1 tat induce expression of IL-4 (which inhibits release of hematopoietic growth factors by other cells) and IL-4 receptors (IL-4R) thereby enhancing the “blunting” effect in lymphocytes159,160 and possibly stromal cells. IL-4 also induces stromal cells to release TGF-β161 and chemokine inhibitors.162 HIV-1 induction of fas ligand (FasL) in mononuclear phagocytes (M) and CD4+ cells results in apoptosis of uninfected CD4+ cells,163,164 but may also result in apoptosis of HPC which are known to express Fas under a variety of conditions. 57-59,165,166 Infection of macrophages also induces release of TNF-α,167,168TGF-β,169,170 and MIP1α.171 CD34+cells exposed to HIV-1 or HIV-1 gp120 release TGF-β,172 but while this has been called an “autocrine” response, it is not known whether the TGF-producing cells are actually progenitor cells, and is not, therefore, known to be a truly autocrine mechanism of HPC inhibition.

Close modal
Monocyte/macrophages.

Apoptosis of lymphoid cells in nodal tissues of patients with HIV-1 infection occurs in cells that are not themselves infected by HIV-1. Priming of the Fas pathway has also been shown in lymphocytes from HIV-1–infected individuals, but cells undergoing apoptosis can be virus free. Badley et al56 have found that HIV-1 induces Fas ligand (FasL) expression in infected monocytes and that this effect may account, indirectly, for apoptotic signals to both lymphocytes and, in our view, possibly progenitor cells (Fig 2). In this report, the investigators argue that this bystander effect, in which HIV-1 induces both Fas and FasL, is a mechanism that can account for lymphocyte depletion during the course of HIV-1 disease. Hematopoietic progenitor cells are also quite susceptible to Fas-induced apoptosis and this particular apoptotic pathway likely accounts for a variety of BM failure states (Fig2).57-59Therefore, it is likely that FasL induction in auxiliary cells may be of substantial relevance, particularly if the Fas pathway is primed by IFN-γ58,60 or chemotherapeutic agents.

Fig. 3.

Accessory cells support adherence and growth of poorly differentiated neoplastic pre-B cells (BL). Dendritic cells,95 CD4+ T cells,34 and other auxiliary cells (Aux)97 infected with HIV-1 or exposed to gp120 or gp16034 release IL-6, a potent mitogen for lymphoma cell growth. Lymphoma cells themselves, both those infected with EBV and EBV, release IL-10 in an autocrine fashion. MVEC infected with HIV-1 express CD40 and bind to lymphoma cell CD40L, a process that activates VCAM-1 expression and subsequent binding with its cognate ligand VLA-4.135Proliferation and survival of lymphoma cells is enhanced following adherence to MVEC. Adherence to MVEC may also facilitate retention of BL cells at sites rich in auxiliary cells for exposure to IL-6.

Fig. 3.

Accessory cells support adherence and growth of poorly differentiated neoplastic pre-B cells (BL). Dendritic cells,95 CD4+ T cells,34 and other auxiliary cells (Aux)97 infected with HIV-1 or exposed to gp120 or gp16034 release IL-6, a potent mitogen for lymphoma cell growth. Lymphoma cells themselves, both those infected with EBV and EBV, release IL-10 in an autocrine fashion. MVEC infected with HIV-1 express CD40 and bind to lymphoma cell CD40L, a process that activates VCAM-1 expression and subsequent binding with its cognate ligand VLA-4.135Proliferation and survival of lymphoma cells is enhanced following adherence to MVEC. Adherence to MVEC may also facilitate retention of BL cells at sites rich in auxiliary cells for exposure to IL-6.

Close modal
BM stromal cells.

Zauli et al33 evaluated the effect of a short-term exposure (2 hours) to two different lymphocytotropic strains of HIV-1 (HIVIIIB and ICR-3) on the survival of a factor-dependent CD34+hematopoietic cell line (TF-1). Although HIV-1–treated TF-1 cells underwent programmed cell death, the response was reversible with optimal doses of IL-3 or GM-CSF or both. Moreover, this group found no signs of productive or latent infection of these cells but did notice that treatment of TF-1 cells with recombinant gp120 plus a polyclonal anti-gp120 antibody, or with anti-CD4 monoclonal antibody (MoAb) plus rabbit anti-mouse IgG, significantly increased the percentage of cells undergoing apoptosis. They reasoned that the apoptotic response was the result of an interaction of gp120 with the CD4 receptor, which was expressed at a low level on the surface of TF-1 cells.

Similarly, in conditions mimicking the steady state in vitro, monocytotropic strains of HIV-1 (Bal, Ada, and JR-FL) did not alter production of TGF-β, TNF-α, MIP-1-α, Steel factor, and IL-6 in long-term BM cultures (LTBMCs).61 Coculture of infected cells with progenitors also failed, at least in one study,61 to alter the number of progenitor cells supported by the stromal layers. The investigators argue that productive and sustained virus replication in the macrophage component of LTBMCs does not significantly alter the profile of major cytokines involved in regulating hematopoiesis at least in the steady state. However, it is important to recognize that this group was unable to infect stromal cells with lymphocytotropic strains,61 likely because the culture conditions were not supportive of microvascular endothelial cell growth. Taking into account the potential importance of stromal MVEC as a reservoir for HIV-1,3,19 these results remained to be confirmed in a long-term system that permits the sustained growth of MVEC. In fact, recently Kohn's group performed studies in which human BM stromal cell cultures were infected ex vivo with a monocytotropic isolate of HIV-1 (JR-FL).52 They reported that stromal cells infected by this isolate fail to normally support the proliferation of hematopoietic progenitor cells but that cells genetically modified to resist HIV-1 infection were fully supportive. Their culture system was not specifically designed to support vascular endothelial cells but did contain 2% MVEC and 2% mononuclear phagocytes. Consequently, mixed marrow stromal cell cultures containing either macrophages or endothelial cells or both are infectable by HIV-1 in vivo and in vitro and such infection reduces the capacity of heterogeneous stromal cells (likely including uninfected ones in the same culture) to support myeloid hematopoiesis. The presence of MVEC in the cultures of this group may account for the differences in the stromal support function described in this study52 compared with other published work.61 

Blunted Inductive Responses in Other Auxiliary Cells

Scadden's group tested the hypothesis that HIV-1 infection of Epo-producing cells might blunt the response of those cells to the inductive effect of hypoxia on Epo gene expression.62 Their studies on the hepatoma cell line Hep3B, an Epo-producing cell line in which hypoxia is a reliable inductive stimulus, show that after direct infection Epo production induced by hypoxia was depressed, potentially by translational suppression of Epo mRNA.62 Clearly, because hepatocyte production of Epo in vivo represents a minor component of the response of humans to hypoxia, the effect of HIV-1 on renal Epo production must be similarly evaluated. However, the potential role of HIV-1, or, more likely, specific viral proteins, in repressing the expression of the epo gene is worth pursuing taking into account the frequent occurrence of anemia in patients with AIDS.

AIDS patients are at increased risk for developing clinically aggressive B-cell non-Hodgkin's lymphomas (NHL).63-66Unlike other AIDS-associated malignancies, the AIDS-associated NHL (AIDS-NHL) develop in every population group at risk for AIDS. Current estimates indicate that 5% to 10% of HIV-1–infected patients develop this life-threatening disease. The etiology of AIDS-NHL is unknown and is likely to be complex. With some interesting exceptions,67,68 the malignant B cells in AIDS-NHL are not directly infected by HIV-1,69-72 suggesting that HIV-1 contributes to B-cell neoplasia via indirect mechanisms. Given the high degree of molecular heterogeneity seen within this group of neoplasms, it is likely that multiple pathways operate individually or in concert within the context of HIV-1 infection to promote lymphomagenesis.

Malignant transformation is the ultimate consequence of sequential genetic changes that occur within a proliferating cell population. Immunocompromised individuals without HIV-1 infection are at increased risk for developing NHL,73 indicating that immunodeficiency per se facilitates transformation. However, NHL arising in immunosuppressed posttransplant patients are generally restricted to the large cell immunoblastic subtype; these patients are not at increased risk for Burkitt's lymphoma.64 This observation suggests that immunosuppression alone is insufficient cause for development of at least the Burkitt's subtype of AIDS-NHL. In addition, AIDS-NHL have a number of distinguishing features including aggressive clinical behavior, extensive extranodal involvement, a high incidence of Epstein-Barr virus (EBV) negativity and a heterogeneous pattern of genetic changes, that suggest the existence of oncogenic mechanisms unique to the HIV-1–infected host. These features are discussed in more detail below.

Distinctive Features of AIDS-NHL

Clinical behavior.

Biggar et al74 recently linked AIDS and cancer registry data for the 1980-1990 decade to evaluate the overall lymphoma risk for persons with AIDS and determined a risk of 348-fold for high-grade lymphomas but only 14-fold for low-grade lymphomas. In another study, over 80% of AIDS-NHL were classified as high-grade tumors while less than 40% of non-AIDS NHL qualified as high-grade neoplasms.75 The aggressive clinical behavior of the AIDS-NHL has necessitated the creation of a separate classification system. Histologically, the majority of systemic AIDS-NHL fall into one of three main categories: (1) high-grade, small noncleaved cell (Burkitt's and Burkitt's-like) lymphomas (SNCCL), (2) high-grade, large cell immunoblastic lymphomas (IBL), and (3) intermediate grade, large noncleaved cell lymphomas (LNCCL). Because of their aggressive clinical behavior, the LNCCL have been functionally classified along with the high-grade IBL as diffuse large cell lymphomas (DLCL). A subtype of “intermediate” lymphomas exhibiting features of both SNCCL and immunoblastic DLCL has also been described.76 The recently described CD30+ anaplastic lymphomas,77 body-cavity–based or primary effusion lymphomas (BCBL/PEL),78,79 and plasmablastic lymphomas (PBL) of the oral cavity80 have also been included in this category, although recent opinion suggests the reclassification of the PEL and PBL, as well as the “intermediate” NHL, as separate pathologic entities.81 Collectively, DLCL comprise about two thirds of the systemic NHL while SNCCL make up approximately one third. T cell, non-T/non-B cell, and low-grade NHL comprise the remainder. The primary central nervous system lymphomas (PCNSL), which account for approximately 20% of all AIDS-NHL, fall almost exclusively into the high-grade immunoblastic subtype of DLCL.

Extranodal involvement.

The extent of extranodal involvement is a characteristic feature of AIDS-NHL. Malignant cells arising in the lymph node may seed to extranodal sites, or the tumor may be exclusively extranodal with no overt involvement of the lymph nodes.82 The most common sites for extranodal involvement include the gastrointestinal tract, liver, BM, and meninges for systemic NHL and perivascular cuffing within the brain parenchyma for the primary CNS NHL.63,83As the name suggests, PEL do not seed from solid tumors but present exclusively in the body cavities as extranodal lymphomatous effusions.

Association with EBV.

While posttransplant NHL arising in immunocompromised patients are invariably EBV-associated, only about half of the AIDS-associated systemic NHL are EBV+.69,70,84-86 Approximately 30% of SNCCL are EBV+ and the transforming antigens EBNA-2 and LMP-1 are not expressed.87 The incidence of EBV among the large cell lymphomas is about 60% to 70%; almost 100% for the immunoblastic subtype where EBNA-2 and LMP-1 are expressed and a lower incidence amongst the LNCCL, which have a latency pattern resembling that of the SNCCL.87 The primary CNS lymphomas are almost exclusively EBV-infected with a latency pattern characterized by expression of EBNA-2 and LMP-1.88 EBV-driven lymphoproliferation in the setting of immunodeficiency is likely to play a central role in the development of EBV+ AIDS-NHL, as it does for posttransplant lymphomas. However, the high frequency of EBV AIDS-NHL indicates that additional factors must influence lymphoma development.

Genetic diversity.

Genetic lesions characteristic of AIDS-NHL are heterogeneous and tend to segregate with certain histologic subtypes. For example, the c-myc gene rearrangement is found in almost all SNCCL, but occurs in only one quarter of DLCL and is absent in the primary CNS lymphomas.89 Inactivation of the tumor suppressor gene p53 occurs in up to 60% of SNCCL but is seen in only a fraction of DLCL,89 while rearrangements of the bcl-6 gene are seen almost exclusively in DLCL.90 The PEL are generally lacking any of these genetic lesions but are exclusively and consistently infected with HHV-8.78,79 The lack of universality of EBV association and the different patterns of EBV gene expression contribute further to the genetic diversity of the AIDS-NHL.

The high degree of clinical and molecular heterogeneity seen within the AIDS-NHL suggests that multiple pathways exist within the HIV-infected host to promote the development of one or another subtype of lymphoma. The unifying feature of these neoplasms, that they all arise abnormally in the setting of AIDS, argues strongly for a general role for HIV-1 infection that encompasses all of these genetically disparate tumors. Thus, it is likely that in the context of the HIV-1–infected host multiple influences cooperate to induce B-cell hyperplasia and facilitate malignant transformation. These influences include: (1) chronic immune stimulation and polyclonal B-cell activation, (2) the presence of a dysregulated cytokine milieu, (3) inadequate tumor surveillance, and (4) infection with potentially oncogenic agents such as EBV and HHV-8.65,72,91 These pathways provide a mechanism for lymphomagenesis without the need to invoke direct infection of the malignant clone. Indeed, it is generally accepted that the malignant B cells in AIDS-NHL are not directly infected by HIV-1.70-72,89 A more detailed description of the host factors that contribute to the development of AIDS-NHL follows.

Host Factors Contributing to the Development of AIDS-NHL

Chronic immune stimulation.

Primary HIV-1 infection is frequently associated with a persistent and generalized lymphadenopathy (PGL), characterized by expansion of lymph node germinal centers in response to the recruitment, proliferation, differentiation, and apoptotic death of antigen-reactive B cells.92 HIV-1 itself, as well as other environmental or self antigens, may contribute to this polyclonal B-cell hyperplasia and hypergammaglobulinemia. Antigen-driven B-cell hyperproliferation would not only increase the risk for genetic accidents and the emergence of transformed B-cell clones, but would contribute to the expansion of such neoplastic clones. In support of a role for chronic antigen stimulation in the development of AIDS-NHL, Riboldi et al93showed that Burkitt's lymphomas derived from AIDS patients produced self-reactive IgM antibodies, and that somatic mutations within the IgM VH segment were reminiscent of those seen in Ig genes from other autoreactive B-cell clones.

Cytokines.

A characteristic feature of AIDS is the existence of a deregulated cytokine network.92 Some of the key cytokines that regulate B-cell growth and differentiation, such as IL-6, IL-9, and IL-10, are produced by CD4+ T cells,34,94 dendritic cells,95,96 and macrophages47,97-99 following HIV-1 infection or in response to viral proteins, suggesting a potential role for these cytokines in B-cell lymphomagenesis. Indeed, high serum levels of IL-6 may be predictive of the development of NHL.100 Once a lymphoma is established, its growth and survival may then be sustained through paracrine and autocrine growth loops.101,102 Interestingly, studies by Benjamin et al103 have shown that autocrine production of IL-10 is a feature of AIDS-associated Burkitt's lymphomas and is not seen in sporadic or endemic cases.

Impaired immune surveillance.

There is a positive correlation between immunodeficiency, as measured by decreasing CD4 counts, and the development of AIDS-NHL.104 This correlation applies particularly to the risk for developing large cell NHL, because SNCCL may develop when immunity is relatively intact.84,100 Independent of CD4 counts, the duration of the immunodeficent state was also found to increase the risk of developing AIDS-NHL.100 This implies that as patients survive longer with improved retroviral treatment, the incidence of AIDS-NHL may increase.105 Impaired immunosurveillance as a risk factor for AIDS-NHL can be explained by an inability of the host to contain EBV-driven B-cell expansions that may precede malignancy, and the defective response of tumor-infiltrating T cells which are known to play an important role in the containment of NHL.106 In addition, generalized immunodeficiency and consequent secondary infections would exacerbate conditions of chronic B-cell stimulation and cytokine deregulation.

Infection with oncogenic viruses.

Viruses that have been causally linked to AIDS-NHL to date include the herpesviruses EBV and, more recently, HHV-8. The oncogenic potential of EBV is demonstrated by the ability of this virus to transform B cells in vitro107 and the capacity of EBV-infected B cells to cause lymphomas in severe combined immunodeficient (SCID) mice.108 However, because EBV infection is not universal amongst the AIDS-NHL, the absolute role of EBV in the development of these neoplasms is unclear. Importantly, EBV infection is highest in primary CNS lymphomas and systemic IBL which arise in the setting of severe immunodeficiency, and lowest in the SNCCL which frequently present while immune competence is relatively preserved. In addition, expression of the transforming antigens EBNA-2 and LMP-1 is restricted to AIDS-NHL associated with advanced immunodeficiency.87,109 This observation strongly suggests that, while the oncogenic potential of EBV is undisputed, the degree to which EBV contributes to AIDS lymphomagenesis depends largely on the degree to which virus replication and gene expression is influenced by the host immune status.

HHV-8, originally designated as Kaposi's sarcoma–associated herpesvirus (KSHV), is the only infectious agent that has been consistently associated with KS lesions in AIDS patients,110,111 and epidemiological studies have supported a causal role for HHV-8 in the pathogenesis of KS.112 HHV-8 infection has also been consistently shown in the PEL subtype of AIDS-NHL which constitute approximately 3% of this group of neoplasms.113 To date HHV-8 sequences have not been identified in the malignant B cells that comprise any of the other subtypes of AIDS-NHL.114 Although HHV-8+ PEL do not occur exclusively in the setting of AIDS, HIV-1 infection significantly increases the risk for developing this type of malignancy.78 The reproducible and selective infection of PEL by HHV-8 provides a compelling argument for a causal role for viral infection in the development of this type of malignancy. Although a tumorogenic role for HHV-8 has yet to be clearly defined, the virus encodes several genes with oncogenic potential as well as a gene encoding a viral homologue of IL-6 (vIL-6).115,116Therefore, it is conceivable that constitutive or deregulated expression of these genes may induce and/or sustain PEL localization and growth.

A Role for Nonmalignant Accessory Cells in AIDS-NHL

While the consequences of a generalized immunodeficiency are no doubt important for neoplastic transformation, the host microenvironment must nurture the expanding cellular clone(s). Although HIV-1 does not influence lymphomagenesis via direct infection of malignant B cells, microenvironments that favor the characteristic features of AIDS-NHL, namely clinically aggressive growth at extranodal sites, may exist uniquely within the HIV-1–infected host. Such microenvironments may in fact be defined by HIV-1 infection of nonmalignant accessory cells. The concept that viral infection of an accessory cell promotes malignant growth of a distinct cell type was recently proposed to explain the growth of plasma cells in multiple myeloma.117 This study established a positive association between multiple myeloma and the infection of BM dendritic cells with HHV-8. Because dendritic cells play an important role in the growth and differentiation of B cells, the investigators proposed that HHV-8–infected dendritic cells contribute to the development of multiple myeloma via expression of viral genes that support the transformation and growth of malignant plasma cells. HIV infection of accessory cells at extranodal locations with the capacity to influence B-cell growth and development may similarly contribute to B-cell transformation in AIDS-NHL and/or homing and growth of malignant clones.

Hematopoietic Stromal Cells Influence B Lymphopoiesis

An essential accessory cell network for normal B-cell lymphopoiesis and homing in vivo, as well as for B-cell growth in vitro, is provided by the BM stroma.118-122 Depending on the conditions under which cells are cultured, BM stromal cultures can be made to be supportive of either lymphopoiesis, as in the Whitlock-Witte or related systems122-125 or myelopoiesis as in the Dexter system.126,127 Clear differences in growth factor production have not been clarified, and while IL-7 production is produced by the majority of cells in stromal cultures that support B-cell growth and differentiation,123 other factors clearly play a role in the support of B cells by marrow stromal elements. Marrow stromal cells support normal and leukemic B-cell progenitor survival and growth128,129 through mechanisms requiring attachment of the cell populations128 via adhesion molecule/ligand interactions such as VCAM-1/VLA-4,130,131and ICAM-1/LFA-1.132 These interactions represent more than mere physical attachment. By attaching to stromal elements, B lymphoma cells induce tyrosine phosphorylation of a number of proteins in stromal cells and, perhaps of more relevance to a juxtacrine mechanism of B-cell growth, B cells induce the release of IL-6.133 

HIV-infected MVEC-enriched stroma supports B-lymphoma cell growth.

Stromal MVEC as well as fibroblasts are effective in supporting B-cell proliferation.134 Indeed, McGinnes et al122reported that MVEC-enriched stroma derived from BM spicules was more effective at supporting B-cell growth than fibroblast-rich aspirate-derived stroma. As previously mentioned, our group and others have shown HIV-1 infection of stromal MVEC within the BM of patients with AIDS and ARC.3,19 More recently, we reported that ex vivo HIV-1 infection of MVEC-enriched stroma isolated from the BM of HIV-1 seronegative lymphoma patients induced the outgrowth and survival of autologous B-lymphoma cells, while such stroma was not supportive of autologous lymphoma growth in the absence of HIV-1 infection.135 In addition, culture of MVEC-enriched stroma isolated from the BM of AIDS patients with B-cell NHL promoted the survival and outgrowth of autologous stromal-dependent malignant B cells. These phenomena were observed for NHL of both the large and small noncleaved B-cell types and included both EBV+ and EBV categories. Consequently, the unifying feature of these stromal cultures was the requirement for HIV-1–infected MVEC. These studies suggest that HIV-1 infection alters the properties of the stromal microenvironment such that it becomes supportive of the outgrowth and survival of B-cell lymphomas. HIV infection of stromal MVEC may thus play a role in the extranodal growth of AIDS-NHL that arise within and/or localize to the BM (Fig 3).

HIV-infected brain MVEC support B-lymphoma cell growth.

HIV-1 is known to infect brain microvascular endothelial cells (brain MVEC) in AIDS patients48,136 and cultured MVEC from normal brain tissue can be productively infected in vitro.49Recent studies from our laboratory have shown that normal brain MVEC cultured in vitro were moderately supportive of the adhesion and growth of B-lymphoma cells added in coculture (Fig 4). Importantly, HIV-1 infection of these brain MVEC dramatically increased the subsequent adhesion and proliferation of cocultured B-lymphoma cells.135 Physical separation of MVEC and B cells, achieved by culturing B-lymphoma cells in transwell filter chambers over HIV-1–infected MVEC monolayers, suggested that enhanced proliferation was dependent on initial MVEC-B cell attachment. Interactions between the adhesion molecule VCAM-1 and the B-cell integrin VLA-4 play a central role in the adhesion of B cells to MVEC and other stromal elements,118,121,122,134,137,138 implicating VCAM-1 as a potential mediator of the enhanced B-lymphoma cell adhesion. Although VCAM-1 expression on brain MVEC is not induced by HIV-1 infection per se,139 expression of the cytokine receptor CD40 by brain MVEC is upregulated after HIV-1 infection of these cells.135 Recent in vitro studies have shown that CD40 is also expressed on dermal and umbilical vein endothelial cells and that CD40 triggering using soluble CD40 ligand (CD40L) increases the constitutive expression of VCAM-1 by these cells.140-142Studies from our laboratory have shown that while CD40 triggering results in a modest induction of VCAM-1 on uninfected brain MVEC, VCAM-1 is significantly induced following CD40 triggering of HIV-1–infected MVEC.135 The ability of CD40 triggering to preferentially induce VCAM-1 on HIV-1–infected MVEC in vitro suggests that a similar mechanism could operate in vivo. While CD40L expression was first identified on activated T cells,143 additional cell types including macrophages, dendritic cells, smooth muscle cells, endothelial cells, and fibroblasts also express CD40L in certain inflammatory states.144,145 In vivo, interaction of HIV-1–infected MVEC with any of these cell types could induce VCAM-1 expression and create a microenvironment conducive to adhesion and growth of malignant B cells. In addition, because CD40L expression by malignant B cells has also been reported,135,146,147CD40L+ B-lymphoma cells could themselves induce the adhesion phenotype. In support of this hypothesis, the adhesion and growth of a CD40L+, VLA-4+ AIDS-SNCCL cell line on HIV-1–infected MVEC is specifically inhibited by blocking functional CD40-CD40L interactions between the cells.135Interactions between CD40 and CD40L may also play a role in the development of other types of malignancies. For example, van den Oord et al148 analyzed malignant melanoma (MM) lesions for expression of CD40 and CD40L and found that 45% of CD40+MM coexpressed CD40L. Interestingly, patients whose tumors expressed both CD40 and CD40L had a poorer prognosis than those expressing CD40 only. The investigators in this study observed that coexpression of CD40 and CD40L usually occurred in the same area of the tumor, and proposed the existence of CD40-CD40L-mediated autocrine growth loops in the vertical growth phase of MM.

Fig. 4.

Clusters of Burkitt's lymphoma cells adhere to and proliferate on HIV-1–infected MVEC from autologous human BM (A) and heterologous human brain (C) but not on uninfected brain MVEC (B). (A) Lymphoma cells and stromal cells in (A) were derived from the BM of an HIV-1 seropositive lymphoma patient. The lymphoma cells were EBV, contained a t(8;14) chromosomal translocation, and proliferated spontaneously in stromal cell cultures that supported MVEC survival. (B) Lymphoma cells from (A) were transferred to pure brain MVEC monolayers. A small percentage of lymphoma cells adhered to brain MVEC, but these brain MVEC did not support sustained adhesion and growth of lymphoma cells. (C) Lymphoma cells from (A) were transferred to pure brain MVEC monolayers infected with HIV-1. Infected MVEC supported the long-term adherence and growth of lymphoma cells.

Fig. 4.

Clusters of Burkitt's lymphoma cells adhere to and proliferate on HIV-1–infected MVEC from autologous human BM (A) and heterologous human brain (C) but not on uninfected brain MVEC (B). (A) Lymphoma cells and stromal cells in (A) were derived from the BM of an HIV-1 seropositive lymphoma patient. The lymphoma cells were EBV, contained a t(8;14) chromosomal translocation, and proliferated spontaneously in stromal cell cultures that supported MVEC survival. (B) Lymphoma cells from (A) were transferred to pure brain MVEC monolayers. A small percentage of lymphoma cells adhered to brain MVEC, but these brain MVEC did not support sustained adhesion and growth of lymphoma cells. (C) Lymphoma cells from (A) were transferred to pure brain MVEC monolayers infected with HIV-1. Infected MVEC supported the long-term adherence and growth of lymphoma cells.

Close modal

Because different lymphoma subtypes express unique classes of regulatory and adhesion molecules,138 HIV-infected brain MVEC likely express additional molecules that enhance MVEC-B lymphoma cell adhesion either directly or via multi-step pathways. For example, the ligand LFA-1 is strongly expressed by certain subsets of NHL138 while its specific counter-receptor, the adhesion molecule ICAM-1, is directly induced on brain MVEC by HIV-1 infection.139 Although our studies to date have not identified any constitutive alteration in cytokine production from HIV-infected brain MVEC, both the physical proximity of B-lymphoma cells to MVEC, as well as the potential for MVEC-B cell adhesion to activate cytokine receptor and cytokine response genes in both cell types, suggest that B cells adherent to HIV-infected MVEC are likely to be optimally responsive to cytokines elaborated from these MVEC, regardless of their absolute amounts.

HIV-1 infection of endothelial cells from a variety of organ systems including the BM,3,19 brain,48,49kidney,51 and liver50 has been reported. The significant correlation between these tissues and extranodal sites targeted for homing and growth of AIDS-NHL strongly suggests that HIV-1–infected endothelial cells function as nonmalignant accessory cells with the capacity to promote the attachment and growth of malignant B cells. The spectrum of phenotypic changes in HIV-1–infected MVEC responsible for enhanced B-lymphoma growth have yet to be fully elucidated, but they will likely be complex. In fact, as would be predicted from the work on uninfected stromal cell/B-cell interactions of LeBien and others,133,134,149 it is very likely that a variety of synergistic mechanisms account for the outgrowths we have seen on HIV-1 infected stromal and brain MVEC (Fig3). Consequently, the inductive changes in adhesion molecule interactions likely represent only the first of many steps in a complex concatenation of induced responses in both accessory and malignant cell elements. However, it is clear that foci of infected endothelial cells, dendritic cells, and macrophages represent a local microenvironment that may, in the setting of AIDS, accommodate a malignant phenotype.

In conclusion, it is generally accepted that in the context of the HIV-1–infected host, the combined consequences of a generalized immunodeficiency, chronic B-cell stimulation, impaired tumor surveillance, and coinfection with oncogenic viruses facilitate the transformation and survival of malignant B cells that are themselves uninfected by HIV-1, and that this mechanism accounts for the high incidence of AIDS-NHL seen within the HIV-1–infected population. In addition, we argue that HIV-1 infection of nonmalignant accessory cells, particularly MVEC, plays an essential role in the homing, growth, and survival of these neoplasms at extranodal sites. While there is still much work to be done to fully elucidate the role of HIV-1 in the development of AIDS-NHL, the clear capacity of HIV-1 to induce changes in the support function of accessory cells for lymphoma cell growth is an essential issue to pursue. Full clarification of the function of the microenvironment in lymphomagenesis will permit the rational design of preventive strategies.

Patients with HIV-1 infection commonly develop pancytopenia, but the causes are heterogeneous and commonly iatrogenic or multifactorial. The most consistent hematopoietic defects that occur in seropositive patients as a result of HIV-1 infection per se include, first, regenerative BM failure in which on-demand hematopoiesis is suppressed, and second, a high frequency of unusually aggressive, extranodal NHLs. It is clear that neither BM failure nor lymphomagenesis results from infection of stem cells or lymphoid or myeloid progenitor cells in vivo. Indeed, it is clear that infection of such cells is not only rare29,150,151 but that the growth and differentiation of the few cells that may be infected is in no way impaired. However, infection of auxiliary cells, particularly macrophages and microvascular endothelial cells, induces a substantial alteration in the supportive function of the hematopoietic stromal tissues such that myeloid hematopoiesis is suppressed and at the same time, primitive lymphoid cell growth is augmented. We argue that a full molecular clarification of these phenomena should lead to opportunities for the rational design of preventive strategies for both regenerative failure and outgrowths of lymphoid neoplasms.

Supported by grants from the National Institutes of Health (DK49887) and the Department of Veterans Affairs.

Address reprint requests to Grover C. Bagby, Jr, MD, Division of Hematology and Medical Oncology, Oregon Health Sciences University, L580, 3181 SW Sam Jackson Park Road, Portland, OR 97201-3098.

1
Mir
N
Costello
C
Luckit
J
Lindley
R
HIV-disease and bone marrow changes: A study of 60 cases.
Eur J Haematol
42
1989
339
2
Costello
C
Haematological abnormalities in human immunodeficiency virus (HIV) disease.
J Clin Pathol
41
1988
711
3
Sun
NCJ
Shapshak
P
Lachant
NA
Hsu
M
Sieger
L
Schmid
P
Beall
G
Imagawa
DT
Bone marrow examination in patients with AIDS and AIDS-related complex (ARC).
Am J Clin Pathol
92
1989
589
4
van Elsacker-Niele AMW: Prevalence of parvovirus B19 infection in patients infected with human immunodeficiency virus. Clin Infect Dis 1997
5
Wickramasinghe
SN
Shiels
S
Bone marrow stromal cell damage in HIV infection.
Clin Lab Haematol
15
1993
236
6
Re
MC
Zauli
G
Furlini
G
Ranieri
S
Monari
P
Ramazzotti
E
La Placa
M
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
129
1993
53
7
Hillyer
CD
Lackey
DA
Villinger
F
Winton
EF
McClure
HM
Ansari
AA
CD34+ and CFU-GM progenitors are significantly decreased in SIVsmm9 infected rhesus macaques with minimal evidence of direct viral infection by polymerase chain reaction.
Am J Hematol
43
1993
274
8
Steinberg
HN
Anderson
J
Crumpacker
CS
Chatis
PA
HIV infection of the BS-1 human stroma cell line: Effect on murine hematopoiesis.
Virology
193
1993
524
9
Dietrich
M
Baumgarte
S
Hutert
F
von Laer
MD
HIV-1 infection of long term bone marrow cultures.
Int Conf AIDS
8
1992
B215
10
(abstr, suppl 1)
LaRussa
VF
Mosca
JD
Kaushal
S
Cutting
MA
Kessler
SW
Reid
T
CD34+ stromal cell precursors are possible targets for HIV.
Blood
82
1993
414a
11
Lunardi-Iskandar
Y
Georgoulias
V
Bertoli
AM
Augery-Bourget
Y
Ammar
A
Vittecoq
D
Rosenbaum
W
Meyer
P
Jasmin
C
Impaired in-vitro proliferation of hemopoietic precursors in HIV-1-infected subjects.
Leuk Res
13
1989
573
12
Dournon
E
Matheron
S
Rozenbaum
W
Gharakhanian
S
Michon
C
Girard
PM
Perronne
C
Salmon
D
De Truchis
P
Leport
C
Effects of zidovudine in 365 consecutive patients with AIDS or AIDS-related complex.
Lancet
2
1988
1297
13
Levine
AM
Wernz
JC
Kaplan
L
Rodman
N
Cohen
P
Metroka
C
Bennett
JM
Rarick
MU
Walsh
C
Kahn
J
Low-dose chemotherapy with central nervous system prophylaxis and zidovudine maintenance in AIDS-related lymphoma. A prospective multi-institutional trial [see comments].
JAMA
266
1991
84
14
Miles
SA
Mitsuyasu
RT
Moreno
J
Baldwin
G
Alton
NK
Souza
L
Glaspy
JA
Combined therapy with recombinant granulocyte colony-stimulating factor and erythropoietin decreases hematologic toxicity from zidovudine.
Blood
77
1991
2109
15
Scadden
DT
Bering
HA
Levine
JD
Bresnahan
J
Evans
L
Epstein
C
Groopman
JE
Granulocyte-macrophage colony-stimulating factor mitigates the neutropenia of combined interferon alpha and zidovudine treatment of acquired immune deficiency syndrome-associated Kaposi's sarcoma.
J Clin Oncol
9
1991
802
16
Pluda
JM
Yarchoan
R
Smith
PD
McAtee
N
Shay
LE
Oette
D
Maha
M
Wahl
SM
Myers
CE
Broder
S
Subcutaneous recombinant granulocyte-macrophage colony-stimulating factor used as a single agent and in an alternating regimen with azidothymidine in leukopenic patients with severe human immunodeficiency virus infection.
Blood
76
1990
463
17
(abstr)
Levine
AM
Cheung
T
Tulpule
A
Huang
J
Testa
M
Preliminary results of AIDS clinical trials group (ACTG) study #149: Phase II trial of ABVD chemotherapy with G-CSF in HIV infected patients with Hodgkin's disease (HD).
J Acquir Immune Defic Syndr Hum Retrovirol
14
1997
A12
18
Bagby GC: Hematopoiesis, in Stamatoyannopoulos G (ed): The Molecular Basis of Hematology. Philadelphia, PA, Saunders, 1993, p 71
19
Moses
AV
Williams
S
Heneveld
ML
Strussenberg
J
Rarick
M
Loveless
M
Bagby
G
Nelson
JA
Human immunodeficiency virus infection of bone marrow endothelium reduces induction of stromal hematopoietic growth factors.
Blood
87
1996
919
20
Freedman
AR
Zhu
H
Levine
JD
Kalams
S
Scadden
DT
Generation of human T lymphocytes from bone marrow CD34+ cells in vitro.
Nat Med
2
1996
46
21
Chelucci
C
Hassan
HJ
Locardi
C
Bulgarini
D
Pelosi
E
Mariani
G
Testa
U
Federico
M
Valtieri
M
Peschle
C
In vitro human immunodeficiency virus-1 infection of purified hematopoietic progenitors in single-cell culture.
Blood
85
1995
1181
22
Schwartz
GN
Kessler
SW
Rothwell
SW
Burrell
LM
Reid
TJ
Meltzer
MS
Wright
DG
Inhibitory effects of HIV-1-infected stromal cell layers on the production of myeloid progenitor cells in human long-term bone marrow cultures [published erratum appears in Exp Hematol 23:181, 1995].
Exp Hematol
22
1994
1288
23
Re
MC
Zauli
G
Gibellini
D
Furlini
G
Ramazzotti
E
Monari
P
Ranieri
S
Capitani
S
La Placa
M
Uninfected haematopoietic progenitor (CD34+) cells purified from the bone marrow of AIDS patients are committed to apoptotic cell death in culture.
AIDS
7
1993
1049
24
Neal
TF
Holland
HK
Baum
CM
Villinger
F
Ansari
AA
Saral
R
Wingard
JR
Fleming
WH
CD34+ progenitor cells from asymptomatic patients are not a major reservoir for human immunodeficiency virus-1.
Blood
86
1995
1749
25
Zauli
G
Re
MC
Visani
G
Furlini
G
Mazza
P
Vignoli
M
La Placa
M
Evidence for a human immunodeficiency virus type 1-mediated suppression of uninfected hematopoietic (CD34+) cells in AIDS patients.
J Infect Dis
166
1992
710
26
Zauli
G
Re
MC
Furlini
G
Giovannini
M
La Placa
M
Human immunodeficiency virus type 1 envelope glycoprotein gp120-mediated killing of human haematopoietic progenitors (CD34+ cells).
J Gen Virol
73
1992
417
27
Molina
JM
Scadden
DT
Sakaguchi
M
Fuller
B
Woon
A
Groopman
JE
Lack of evidence for infection of or effect on growth of hematopoietic progenitor cells after in vivo or in vitro exposure to human immunodeficiency virus.
Blood
76
1990
2476
28
Davis
BR
Schwartz
DH
Marx
JC
Johnson
CE
Berry
JM
Lyding
J
Merigan
TC
Zander
A
Absent or rare human immunodeficiency virus infection of bone marrow stem/progenitor cells in vivo.
J Virol
65
1991
1985
29
De Luca
A
Teofili
L
Antinori
A
Iovino
MS
Mencarini
P
Visconti
E
Tamburrini
E
Leone
G
Ortona
L
Haemopoietic CD34+ progenitor cells are not infected by HIV-1 in vivo but show impaired clonogenesis.
Br J Haematol
85
1993
20
30
Bagnara
GP
Zauli
G
Giovannini
M
Re
MC
Furlini
G
La Placa
M
Early loss of circulating hemopoietic progenitors in HIV-1-infected subjects.
Exp Hematol
18
1990
426
31
Slobod
KS
Bennett
TA
Freiden
PJ
Kechli
AM
Howlett
N
Flynn
PM
Head
DR
Srivastava
DK
Boyett
JM
Brenner
MK
Garcia
JV
Mobilization of CD34+ progenitor cells by granulocyte colony-stimulating factor in human immunodeficiency virus type 1-infected adults.
Blood
88
1996
3329
32
Miles
SA
Mitsuyasu
RT
Lee
K
Moreno
J
Alton
K
Egrie
JC
Souza
L
Glaspy
JA
Recombinant human granulocyte colony-stimulating factor increases circulating burst forming unit-erythron and red blood cell production in patients with severe human immunodeficiency virus infection.
Blood
75
1990
2137
33
Zauli
G
Vitale
M
Re
MC
Furlini
G
Zamai
L
Falcieri
E
Gibellini
D
Visani
G
Davis
BR
Capitani
S
In vitro exposure to human immunodeficiency virus type 1 induces apoptotic cell death of the factor-dependent TF-1 hematopoietic cell line.
Blood
83
1994
167
34
Oyaizu
N
Chirmule
N
Ohnishi
Y
Kalyanaraman
VS
Pahwa
S
Human immunodeficiency virus type 1 envelope glycoproteins gp120 and gp160 induce interleukin-6 production in CD4+ T-cell clones.
J Virol
65
1991
6277
35
Shearer
GM
Roilides
E
Pizzo
PA
Clerici
M
CD4+ T helper cell function is actively suppressed in HIV infection.
Int Conf AIDS
7
1991
61
36
Luria
S
Chambers
I
Berg
P
Expression of the type 1 human immunodeficiency virus Nef protein in T cells prevents antigen receptor-mediated induction of interleukin 2 mRNA.
Proc Natl Acad Sci USA
88
1991
5326
37
Smith
BL
Krushelnycky
BW
Mochly-Rosen
D
Berg
P
The HIV Nef protein associates with protein kinase C theta.
J Biol Chem
271
1996
16753
38
Baier
G
Baier-Bitterlich
G
Meller
N
Coggeshall
KM
Giampa
L
Telford
D
Isakov
N
Altman
A
Expression and biochemical characterization of human protein kinase C-theta.
Eur J Biochem
225
1994
195
39
Monks
CRF
Kupfer
H
Tamir
E
Barlow
A
Kupfer
A
Selective modulation of protein kinase C-q during T-cell activation.
Nature
385
1997
83
40
Stella
CC
Ganser
A
Hoelzer
D
Defective in vitro growth of the hemopoietic progenitor cells in the acquired immunodeficiency syndrome.
J Clin Invest
80
1987
286
41
Molina
JM
Scadden
DT
Amirault
C
Woon
A
Vannier
E
Dinarello
CA
Groopman
JE
Human immunodeficiency virus does not induce interleukin-1, interleukin-6, or tumor necrosis factor in mononuclear cells.
J Virol
64
1990
2901
42
Chehimi
J
Starr
SE
Frank
I
D'Andrea
A
Ma
X
MacGregor
RR
Sennelier
J
Trinchieri
G
Impaired interleukin 12 production in human immunodeficiency virus-infected patients.
J Exp Med
179
1994
1361
43
Sperber
K
Hamrang
G
Louie
MJ
Kalb
T
Banerjee
R
Choi
HS
Paronetto
F
Mayer
L
Progressive impairment of monocytic function in HIV-1-infected human macrophage hybridomas.
AIDS Res Hum Retroviruses
9
1993
657
44
Bagby
GC
Interleukin 1 and hematopoiesis.
Blood Rev
3
1989
152
45
Tripp
CS
Wolf
SF
Unanue
ER
Interleukin 12 and tumor necrosis factor alpha are costimulators of interferon gamma production by natural killer cells in severe combined immunodeficiency mice with listeriosis, and interleukin 10 is a physiologic antagonist.
Proc Natl Acad Sci USA
90
1993
3725
46
Wu
C-Y
Demeure
C
Kiniwa
M
Gately
M
Delespesse
G
IL-12 induces the production of IFN-gamma by neonatal human CD4 T cells.
J Immunol
151
1993
1938
47
Esser
R
Glienke
W
Von Briesen
H
Rubsamen-Waigmann
H
Andreesen
R
Differential regulation of proinflammatory and hematopoietic cytokines in human macrophages after infection with human immunodeficiency virus.
Blood
88
1996
3474
48
Wiley
CA
Schrier
RD
Nelson
JA
Lampert
PW
Oldstone
MB
Cellular localization of human immunodeficiency virus infection within the brains of acquired immune deficiency syndrome patients.
Proc Natl Acad Sci USA
83
1986
7089
49
Moses
AV
Bloom
FE
Pauza
CD
Nelson
JA
HIV infection of human brain capillary endothelial cells occurs via a CD4/GAL C-independent mechanism.
Proc Natl Acad Sci USA
90
1993
10474
50
Steffan
AM
Lafon
ME
Gendrault
JL
Schweitzer
C
Royer
C
Jaeck
D
Arnaud
JP
Schmitt
MP
Aubertin
AM
Kirn
A
Primary cultures of endothelial cells from the human liver sinusoid are permissive for human immunodeficiency virus type 1.
Proc Natl Acad Sci USA
89
1992
1582
51
Green
DF
Resnick
L
Bourgoignie
JJ
HIV infects glomerular endothelial and mesangial but not epithelial cells in vitro.
Kidney Int
41
1992
956
52
Bahner
I
Kearns
K
Coutinho
S
Leonard
EH
Kohn
DB
Infection of human marrow stroma by human immunodeficiency virus-1 (HIV-1) is both required and sufficient for HIV-1-induced hematopoietic suppression in vitro: Demonstration by gene modification of primary human stroma.
Blood
90
1997
1787
53
Bagby
GC
T-lymphocytes which inhibit granulopoiesis in two neutropenic patients are of the cytotoxic suppressor (OKT3+, OKT8+) subset.
J Clin Invest
68
1981
1597
54
Bagby
GC
Lawrence
JH
Neerhout
RC
Autoimmune granulopoietic failure: Identification of T-lymphocyte-mediated inhibition of granulopoiesis and prednisone responsiveness using in vitro techniques.
N Engl J Med
309
1983
1073
55
Geissler
RG
Rossol
R
Mentzel
U
Ottmann
OG
Klein
AS
Gute
P
Helm
EB
Hoelzer
D
Ganser
A
Gamma delta-T cell-receptor-positive lymphocytes inhibit human hematopoietic progenitor cell growth in HIV type 1-infected patients.
AIDS Res Hum Retroviruses
12
1996
577
56
Badley
AD
McElhinny
JA
Leibson
PJ
Lynch
DH
Alderson
MR
Paya
CV
Upregulation of Fas ligand expression by human immunodeficiency virus in human macrophages mediates apoptosis of uninfected T lymphocytes.
J Virol
70
1996
199
57
Nagafuji
K
Shibuya
T
Harada
M
Mizuno
S
Takenaka
K
Miyamoto
T
Okamura
T
Gondo
H
Niho
Y
Functional expression of Fas antigen (CD95) on hematopoietic progenitor cells.
Blood
86
1995
883
58
Maciejewski
J
Selleri
C
Anderson
S
Young
NS
Fas antigen expression on CD34+ human marrow cells is induced by interferon gamma and tumor necrosis factor alpha and potentiates cytokine-mediated hematopoietic suppression in vitro.
Blood
85
1995
3183
59
Maciejewski
JP
Selleri
C
Sato
T
Anderson
S
Young
NS
Increased expression of Fas antigen on bone marrow CD34+ cells of patients with aplastic anaemia.
Br J Haematol
91
1995
245
60
Rathbun
RK
Faulkner
GR
Ostroski
MH
Christianson
TA
Hughes
G
Jones
G
Cahn
R
Maziarz
RT
Royle
G
Keeble
W
Heinrich
MC
Grompe
M
Tower
PA
Bagby
GC
Inactivation of the Fanconi anemia group C (FAC) gene augments interferon-gamma-induced apoptotic responses in hematopoietic cells.
Blood
90
1997
974
61
Marandin
A
Canque
B
Coulombel
L
Gluckman
JC
Vainchenker
W
Louache
F
In vitro infection of bone marrow-adherent cells by human immunodeficiency virus type 1 (HIV-1) does not alter their ability to support hematopoiesis.
Virology
213
1995
245
62
Wang
Z
Goldberg
MA
Scadden
DT
HIV-1 suppresses erythropoietin production in vitro.
Exp Hematol
21
1993
683
63
Ziegler
JL
Drew
WL
Miner
RC
Mintz
L
Rosenbaum
E
Gershow
J
Lennette
ET
Greenspan
J
Shillitoe
E
Beckstead
J
Casavant
C
Yamamoto
K
Outbreak of Burkitt's-like lymphoma in homosexual men.
Lancet
2
1982
631
64
Beral
V
Peterman
T
Berkelman
R
Jaffe
H
AIDS-associated non-Hodgkin lymphoma [see comments].
Lancet
337
1991
805
65
Karp
JE
Broder
S
Acquired immunodeficiency syndrome and non-Hodgkin's lymphomas.
Cancer Res
51
1991
4743
66
Levine
AM
AIDS-related malignancies: The emerging epidemic.
J Natl Cancer Inst
85
1993
1382
67
Astrin
SM
Schattner
E
Laurence
J
Lebman
RI
Rodriguez-Alfageme
C
Does HIV infection of B lymphocytes initiate AIDS lymphoma? Detection by PCR of viral sequences in lymphoma tissue.
Curr Top Microbiol Immunol
182
1992
399
68
Prevot
S
Raphael
M
Fournier
JG
Diebold
J
Detection by in situ hybridization of HIV and c-myc RNA in tumour cells of AIDS-related B-cell lymphomas.
Histopathology
22
1993
151
69
Subar
M
Neri
A
Inghirami
G
Knowles
DM
Dalla-Favera
R
Frequent c-myc oncogene activation and infrequent presence of Epstein-Barr virus genome in AIDS-associated lymphoma.
Blood
72
1988
667
70
Hamilton-Dutoit
SJ
Pallesen
G
Franzmann
MB
Karkov
J
Black
F
Skinhoj
P
Pedersen
C
AIDS-related lymphoma. Histopathology, immunophenotype, and association with Epstein-Barr virus as demonstrated by in situ nucleic acid hybridization.
Am J Pathol
138
1991
149
71
Groopman
JE
Sullivan
JL
Mulder
C
Ginsburg
D
Orkin
SH
O'Hara
CJ
Falchuk
K
Wong-Staal
F
Gallo
RC
Pathogenesis of B cell lymphoma in a patient with AIDS.
Blood
67
1986
612
72
Pelicci
PG
Knowles
DM
2d
Arlin
ZA
Wieczorek
R
Luciw
P
Dina
D
Basilico
C
Dalla-Favera
R
Multiple monoclonal B cell expansions and c-myc oncogene rearrangements in acquired immune deficiency syndrome-related lymphoproliferative disorders. Implications for lymphomagenesis.
J Exp Med
164
1986
2049
73
(suppl 19)
Kinlen
L
Immunosuppressive therapy and acquired immunological disorders.
Cancer Res
52
1992
5474s
74
(abstr)
Biggar
RJ
Rosenberg
PS
Cote
T
Goedert
JJ
Non Hodgkin's lymphoma in AIDS: findings from linking AIDS and cancer registries.
J Acquir Immune Defic Syndr Hum Retrovirol
14
1997
A43
75
Levine
AM
Shibata
D
Sullivan-Halley
J
Nathwani
B
Brynes
R
Slovak
ML
Mahterian
S
Riley
CL
Weiss
L
Levine
PH
Epidemiological and biological study of acquired immunodeficiency syndrome-related lymphoma in the County of Los Angeles: Preliminary results.
Cancer Res
52
1992
5482s
76
Delecluse
HJ
Raphael
M
Magaud
JP
Felman
P
Alsamad
IA
Bornkamm
GW
Lenoir
GM
Variable morphology of human immunodeficiency virus-associated lymphomas with c-myc rearrangements. The French Study Group of Pathology for Human Immunodeficiency Virus-Associated Tumors, I.
Blood
82
1993
552
77
Chadburn
A
Cesarman
E
Jagirdar
J
Subar
M
Mir
RN
Knowles
DM
CD30 (Ki-1) positive anaplastic large cell lymphomas in individuals infected with the human immunodeficiency virus.
Cancer
72
1993
3078
78
Carbone
A
Gloghini
A
Vaccher
E
Zagonel
V
Pastore
C
Dalla Palma
P
Branz
F
Saglio
G
Volpe
R
Tirelli
U
Gaidano
G
Kaposi's sarcoma-associated herpesvirus DNA sequences in AIDS-related and AIDS-unrelated lymphomatous effusions.
Br J Haematol
94
1996
533
79
Cesarman
E
Chang
Y
Moore
PS
Said
JW
Knowles
DM
Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas [see comments].
N Engl J Med
332
1995
1186
80
Delecluse
HJ
Anagnostopoulos
I
Dallenbach
F
Hummel
M
Marafioti
T
Schneider
U
Huhn
D
Schmidt-Westhausen
A
Reichart
PA
Gross
U
Stein
H
Plasmablastic lymphomas of the oral cavity: A new entity associated with the human immunodeficiency virus infection.
Blood
89
1997
1413
81
Carbone
A
Gloghini
A
Canzonieri
V
Tirelli
U
AIDS-related extranodal non-Hodgkin's lymphomas with plasma cell differentiation.
Blood
90
1997
1337
82
Raphael
M
Gentilhomme
O
Tulliez
M
Byron
PA
Diebold
J
Histopathologic features of high-grade non-Hodgkin's lymphomas in acquired immunodeficiency syndrome. The French Study Group of Pathology for Human Immunodeficiency Virus-Associated Tumors.
Arch Pathol Lab Med
115
1991
15
83
Lowenthal
DA
Straus
DJ
Campbell
SW
Gold
JW
Clarkson
BD
Koziner
B
AIDS-related lymphoid neoplasia. The Memorial Hospital experience.
Cancer
61
1988
2325
84
Pedersen
C
Gerstoft
J
Lundgren
JD
Skinhoj
P
Bottzauw
J
Geisler
C
Hamilton-Dutoit
SJ
Thorsen
S
Lisse
I
Ralfkiaer
E
HIV-associated lymphoma: Histopathology and association with Epstein-Barr virus genome related to clinical, immunological and prognostic features.
Eur J Cancer
27
1991
1416
85
Shibata
D
Weiss
LM
Hernandez
AM
Nathwani
BN
Bernstein
L
Levine
AM
Epstein-Barr virus-associated non-Hodgkin's lymphoma in patients infected with the human immunodeficiency virus.
Blood
81
1993
2102
86
Shiramizu
B
Herndier
B
Meeker
T
Kaplan
L
McGrath
M
Molecular and immunophenotypic characterization of AIDS-associated, Epstein-Barr virus-negative, polyclonal lymphoma [see comments].
J Clin Oncol
10
1992
383
87
Carbone
A
Tirelli
U
Gloghini
A
Volpe
R
Boiocchi
M
Human immunodeficiency virus-associated systemic lymphomas may be subdivided into two main groups according to Epstein-Barr viral latent gene expression.
J Clin Oncol
11
1993
1674
88
Camilleri-Broet
S
Davi
F
Feuillard
J
Bourgeois
C
Seilhean
D
Hauw
JJ
Raphael
M
High expression of latent membrane protein 1 of Epstein-Barr virus and BCL-2 oncoprotein in acquired immunodeficiency syndrome-related primary brain lymphomas.
Blood
86
1995
432
89
Ballerini
P
Gaidano
G
Gong
JZ
Tassi
V
Saglio
G
Knowles
DM
Dalla-Favera
R
Multiple genetic lesions in acquired immunodeficiency syndrome-related non-Hodgkin's lymphoma.
Blood
81
1993
166
90
Gaidano
G
Lo Coco
F
Ye
BH
Shibata
D
Levine
AM
Knowles
DM
Dalla-Favera
R
Rearrangements of the BCL-6 gene in acquired immunodeficiency syndrome-associated non-Hodgkin's lymphoma: Association with diffuse large-cell subtype.
Blood
84
1994
397
91
Boyle
MJ
Goldstein
DA
Frazer
IH
Sculley
TB
Managing HIV. Part 3: Mechanisms of disease. 3.6. How HIV promotes malignancies.
Med J Aust
164
1996
230
92
Fauci
AS
Schnittman
SM
Poli
G
Koenig
S
Pantaleo
G
Immunopathogenic mechanisms in human immunodeficiency virus (HIV) infection.
Ann Int Med
114
1991
678
93
Riboldi
P
Gaidano
G
Schettino
EW
Steger
TG
Knowles
DM
Dalla-Favera
R
Casali
P
Two acquired immunodeficiency syndrome-associated Burkitt's lymphomas produce specific anti-i IgM cold agglutinins using somatically mutated VH4-21 segments.
Blood
83
1994
2952
94
Chirmule
N
Kalyanaraman
VS
Lederman
S
Oyaizu
N
Yagura
H
Yellin
MJ
Chess
L
Pahwa
S
HIV-gp 160-induced T cell-dependent B cell differentiation. Role of T cell-B cell activation molecule and IL-6.
J Immunol
150
1993
2478
95
Ghanekar
S
Zheng
L
Logar
A
Navratil
J
Borowski
L
Gupta
P
Rinaldo
C
Cytokine expression by human peripheral blood dendritic cells stimulated in vitro with HIV-1 and herpes simplex virus.
J Immunol
157
1996
4028
96
Ludewig
B
Gelderblom
HR
Becker
Y
Schäfer
A
Pauli
G
Transmission of HIV-1 from productively infected mature Langerhans cells to primary CD4+ T lymphocytes results in altered T cell responses with enhanced production of IFN-gamma and IL-10.
Virology
215
1996
51
97
Clouse
KA
Cosentino
LM
Weih
KA
Pyle
SW
Robbins
PB
Hochstein
HD
Natarajan
V
Farrar
WL
The HIV-1 gp120 envelope protein has the intrinsic capacity to stimulate monokine secretion.
J Immunol
147
1991
2892
98
Trentin
L
Garbisa
S
Zambello
R
Agostini
C
Caenazzo
C
Di Francesco
C
Cipriani
A
Francavilla
E
Semenzato
G
Spontaneous production of interleukin-6 by alveolar macrophages from human immunodeficiency virus type 1-infected patients.
J Infect Dis
166
1992
731
99
Gan
HX
Ruef
C
Hall
BF
Tobin
E
Remold
HG
Mellors
JW
Interleukin-6 expression in primary macrophages infected with human immunodeficiency virus-1 (HIV-1).
AIDS Res Hum Retroviruses
7
1991
671
100
Pluda
JM
Venzon
DJ
Tosato
G
Lietzau
J
Wyvill
K
Nelson
DL
Jaffe
ES
Karp
JE
Broder
S
Yarchoan
R
Parameters affecting the development of non-Hodgkin's lymphoma in patients with severe human immunodeficiency virus infection receiving antiretroviral therapy.
J Clin Oncol
11
1993
1099
101
Emilie
D
Coumbaras
J
Raphael
M
Devergne
O
Delecluse
HJ
Gisselbrecht
C
Michiels
JF
Van Damme
J
Taga
T
Kishimoto
T
Interleukin-6 production in high-grade B lymphomas: Correlation with the presence of malignant immunoblasts in acquired immunodeficiency syndrome and in human immunodeficiency virus-seronegative patients.
Blood
80
1992
498
102
Emilie
D
Touitou
R
Raphael
M
Peuchmaur
M
Devergnee
O
Rea
D
Coumbraras
J
Crevon
MC
Edelman
L
Joab
I
In vivo production of interleukin-10 by malignant cells in AIDS lymphomas.
Eur J Immunol
22
1992
2937
103
Benjamin
D
Knobloch
TJ
Dayton
MA
Human B-cell interleukin-10: B-cell lines derived from patients with acquired immunodeficiency syndrome and Burkitt's lymphoma constitutively secrete large quantities of interleukin-10.
Blood
80
1992
1289
104
Levine
AM
Sullivan-Halley
J
Pike
MC
Rarick
MU
Loureiro
C
Bernstein-Singer
M
Willson
E
Brynes
R
Parker
J
Rasheed
S
Human immunodeficiency virus-related lymphoma. Prognostic factors predictive of survival [see comments].
Cancer
68
1991
2466
105
Kaplan LD: The lymphoma project report. Current issues in research and treatment of AIDS-associated lymphoma. Pamphlet published by The Treatment Action Group, New York, NY, 1995
106
Ramsay
AD
Smith
WJ
Isaacson
PG
T-cell-rich B-cell lymphoma.
Am J Surg Pathol
12
1988
433
107
Kieff E, Liebowitz D: Oncogenesis by herpesviruses, in Weinberg RA (ed): Oncogenes and the Molecular Origins of Cancer. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory, 1989, p 259
108
Rowe
M
Young
LS
Crocker
J
Stokes
H
Henderson
S
Rickinson
AB
Epstein-Barr virus (EBV)-associated lymphoproliferative disease in the SCID mouse model: Implications for the pathogenesis of EBV-positive lymphomas in man.
J Exp Med
173
1991
147
109
Hamilton-Dutoit
SJ
Raphael
M
Audouin
J
Diebold
J
Lisse
I
Pedersen
C
Oksenhendler
E
Marelle
L
Pallesen
G
In situ demonstration of Epstein-Barr virus small RNAs (EBER 1) in acquired immunodeficiency syndrome-related lymphomas: correlation with tumor morphology and primary site.
Blood
82
1993
619
110
Chang
Y
Cesarman
E
Pessin
MS
Lee
F
Culpepper
J
Knowles
DM
Moore
PS
Identification of herpesvirus-like DNA sequences in AIDS- associated Kaposi's sarcoma.
Science
266
1994
1865
111
Moore
PS
Chang
Y
Detection of herpesvirus-like DNA sequences in Kaposi's sarcoma in patients with and without HIV infection [see comments].
N Engl J Med
332
1995
1181
112
Whitby
D
Howard
MR
Tenant-Flowers
M
Brink
NS
Copas
A
Boshoff
C
Hatzioannou
T
Suggett
FE
Aldam
DM
Denton
AS
Detection of Kaposi sarcoma associated herpesvirus in peripheral blood of HIV-infected individuals and progression to Kaposi's sarcoma [see comments].
Lancet
346
1995
799
113
Lipkin
M
Higgins
P
Biological markers of cell proliferation and differentiation in human gastrointestinal diseases.
Adv Cancer Res
50
1988
1
114
Gaidano
G
Pastore
C
Gloghini
A
Cusini
M
Nomdedeu
J
Volpe
G
Capello
D
Vaccher
E
Bordes
R
Tirelli
U
Saglio
G
Carbone
A
Distribution of human herpesvirus-8 sequences throughout the spectrum of AIDS-related neoplasia.
AIDS
10
1996
941
115
Cesarman
E
Nador
RG
Bai
F
Bohenzky
RA
Russo
JJ
Moore
PS
Chang
Y
Knowles
DM
Kaposi's sarcoma-associated herpesvirus contains G protein-coupled receptor and cyclin D homologs which are expressed in Kaposi's sarcoma and malignant lymphoma.
J Virol
70
1996
8218
116
Moore
PS
Boshoff
C
Weiss
RA
Chang
Y
Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV.
Science
274
1996
1739
117
Rettig
MB
Ma
HJ
Vescio
RA
Pold
M
Schiller
G
Belson
D
Savage
A
Nishikubo
C
Wu
C
Fraser
J
Said
JW
Berenson
JR
Kaposi's sarcoma-associated herpesvirus infection of bone marrow dendritic cells from multiple myeloma patients [see comments].
Science
276
1997
1851
118
Tang
J
Scott
G
Ryan
DH
Subpopulations of bone marrow fibroblasts support VLA-4-mediated migration of B-cell precursors.
Blood
82
1993
3415
119
Jacobsen
K
Osmond
DG
Microenvironmental organization and stromal cell associations of B lymphocyte precursor cells in mouse bone marrow.
Eur J Immunol
20
1990
2395
120
Wolf
ML
Buckley
JA
Goldfarb
A
Law
CL
LeBien
TW
Development of a bone marrow culture for maintenance and growth of normal human B cell precursors.
J Immunol
147
1991
3324
121
Simmons
PJ
Masinovsky
B
Longenecker
BM
Berenson
R
Torok-Storb
B
Gallatin
WM
Vascular cell adhesion molecule-1 expressed by bone marrow stromal cells mediates the binding of hematopoietic progenitor cells.
Blood
80
1992
388
122
McGinnes
K
Quesniaux
V
Hitzler
J
Paige
C
Human B-lymphopoiesis is supported by bone marrow-derived stromal cells.
Exp Hematol
19
1991
294
123
Witte
PL
Frantsve
LM
Hergott
M
Rahbe
SM
Cytokine production and heterogeneity of primary stromal cells that support B lymphopoiesis.
Eur J Immunol
23
1993
1809
124
Whitlock
CA
Witte
ON
Long-term culture of murine bone marrow precursors of B lymphocytes.
Methods Enzymol
150
1987
275
125
Whitlock
CA
Witte
ON
Long-term culture of B lymphocytes and their precursors from murine bone marrow.
Proc Natl Acad Sci USA
79
1982
3608
126
Dexter
TM
Haemopoiesis in long-term bone marrow cultures. A review.
Acta Haematol
62
1979
299
127
Dexter
TM
Allen
TD
Lajtha
LG
Conditions controlling the proliferation of haemopoietic stem cells in vitro.
J Cell Physiol
91
1977
335
128
Manabe
A
Murti
KG
Coustan-Smith
E
Kumagai
M
Behm
FG
Raimondi
SC
Campana
D
Adhesion-dependent survival of normal and leukemic human B lymphoblasts on bone marrow stromal cells.
Blood
83
1994
758
129
Panayiotidis
P
Jones
D
Ganeshaguru
K
Foroni
L
Hoffbrand
AV
Human bone marrow stromal cells prevent apoptosis and support the survival of chronic lymphocytic leukaemia cells in vitro.
Br J Haematol
92
1996
97
130
Miyake
K
Weissman
IL
Greenberger
JS
Kincade
PW
Evidence for a role of the integrin VLA-4 in lympho-hemopoiesis.
J Exp Med
173
1991
599
131
Juneja
HS
Schmalsteig
FC
Lee
S
Chen
J
Vascular cell adhesion molecule-1 and VLA-4 are obligatory adhesion proteins in the heterotypic adherence between human leukemia/lymphoma cells and marrow stromal cells.
Exp Hematol
21
1993
444
132
Koopman
G
Keehnen
RM
Lindhout
E
Newman
W
Shimizu
Y
van Seventer
GA
de Groot
C
Pals
ST
Adhesion through the LFA-1 (CD11a/CD18)-ICAM-1 (CD54) and the VLA-4 (CD49d)-VCAM-1 (CD106) pathways prevents apoptosis of germinal center B cells.
J Immunol
152
1994
3760
133
Jarvis
LJ
LeBien
TW
Stimulation of human bone marrow stromal cell tyrosine kinases and IL-6 production by contact with B lymphocytes.
J Immunol
155
1995
2359
134
Dittel
BN
McCarthy
JB
Wayner
EA
LeBien
TW
Regulation of human B-cell precursor adhesion to bone marrow stromal cells by cytokines that exert opposing effects on the expression of vascular cell adhesion molecule-1 (VCAM-1).
Blood
81
1993
2272
135
Moses
AV
Williams
SE
Strussenberg
JG
Ostroski
ML
Bagby
GC
Nelson
J
HIV-1 induction of CD40 on endothelial cells promotes the outgrowth of AIDS-associated B cell lymphomas.
Nature Med
3
1997
1242
136
Bagasra
O
Lavi
E
Bobroski
L
Khalili
K
Pestaner
JP
Tawadros
R
Pomerantz
RJ
Cellular reservoirs of HIV-1 in the central nervous system of infected individuals: Identification by the combination of in situ polymerase chain reaction and immunohistochemistry.
AIDS
10
1996
573
137
Ryan
DH
Nuccie
BL
Abboud
CN
Winslow
JM
Vascular cell adhesion molecule-1 and the integrin VLA-4 mediate adhesion of human B cell precursors to cultured bone marrow adherent cells.
J Clin Invest
88
1991
995
138
Freedman
AS
Expression and function of adhesion receptors on normal B cells and B cell non-Hodgkin's lymphomas.
Semin Hematol
30
1993
318
139
Moses
AV
Nelson
JA
HIV infection of human brain capillary endothelial cells—Implications for AIDS dementia.
Adv Neuroimmunol
4
1994
239
140
Hollenbaugh
D
Mischel-Petty
N
Edwards
CP
Simon
JC
Denfeld
RW
Kiener
PA
Aruffo
A
Expression of functional CD40 by vascular endothelial cells.
J Exp Med
182
1995
33
141
Karmann
K
Hughes
CC
Schechner
J
Fanslow
WC
Pober
JS
CD40 on human endothelial cells: Inducibility by cytokines and functional regulation of adhesion molecule expression.
Proc Natl Acad Sci USA
92
1995
4342
142
Yellin
MJ
Brett
J
Baum
D
Matsushima
A
Szabolcs
M
Stern
D
Chess
L
Functional interactions of T cells with endothelial cells: The role of CD40L-CD40-mediated signals.
J Exp Med
182
1995
1857
143
Noelle
RJ
Roy
M
Shepherd
DM
Stamenkovic
I
Ledbetter
JA
Aruffo
A
A 39-kDa protein on activated helper T cells binds CD40 and transduces the signal for cognate activation of B cells.
Proc Natl Acad Sci USA
89
1992
6550
144
Cocks
BG
De Waal Malefyt
R
Galizzi
JP
De Vries
JE
Aversa
G
IL-13 induces proliferation and differentiation of human B cells activated by the CD40 ligand.
Int Immunol
5
1993
657
145
Mach
F
Schonbeck
U
Sukhova
GK
Bourcier
T
Bonnefoy
JY
Pober
JS
Libby
P
Functional CD40 ligand is expressed on human vascular endothelial cells, smooth muscle cells, and macrophages: Implications for CD40-CD40 ligand signaling in atherosclerosis.
Proc Natl Acad Sci USA
94
1997
1931
146
Grammer
AC
Bergman
MC
Miura
Y
Fujita
K
Davis
LS
Lipsky
PE
The CD40 ligand expressed by human B cell costimulates B cell responses.
J Immunol
154
1995
4996
147
Pinchuk
LM
Klaus
SJ
Magaletti
DM
Pinchuk
GV
Norsen
JP
Clark
EA
Functional CD40 ligand expressed by human blood dendritic cells is up-regulated by CD40 ligation.
J Immunol
157
1996
4363
148
van den Oord
JJ
Maes
A
Stas
M
Nuyts
J
Battocchio
S
Kasran
A
Garmyn
M
De Wever
I
De Wolf-Peeters
C
CD40 is a prognostic marker in primary cutaneous malignant melanoma.
Am J Pathol
149
1996
1953
149
Larson
AW
LeBien
TW
Cross-linking CD40 on human B cell precursors inhibits or enhances growth depending on the stage of development and the IL costimulus.
J Immunol
153
1994
584
150
Stanley
SK
Kessler
SW
Justement
JS
Schnittman
SM
Greenhouse
JJ
Brown
CC
Musongela
L
Musey
K
Kapita
B
Fauci
AS
CD34+ bone marrow cells are infected with HIV in a subset of seropositive individuals.
J Immunol
149
1992
689
151
Kearns
K
Bahner
I
Bauer
G
Wei
S
Valdez
P
Wheeler
S
Woods
L
Miller
R
Casciato
D
Galpin
J
Church
J
Kohn
DB
Suitability of bone marrow from HIV-1-infected donors for retrovirus-mediated gene transfer.
Human Gene Ther
8
1997
301
152
Blazevic
V
Heino
M
Lagerstedt
A
Ranki
A
Krohn
KJ
Interleukin-10 gene expression induced by HIV-1 Tat and Rev in the cells of HIV-1 infected individuals.
J Acquir Immune Defic Syndr Hum Retrovirol
13
1996
208
153
Takeshita
S
Breen
EC
Ivashchenko
M
Nishanian
PG
Kishimoto
T
Vredevoe
DL
Martinez-Maza
O
Induction of IL-6 and IL-10 production by recombinant HIV-1 envelope glycoprotein 41 (gp41) in the THP-1 human monocytic cell line.
Cell Immunol
165
1995
234
154
De Waal Malefyt
R
Abrams
J
Bennett
B
Figdor
CG
De Vries
JE
Interleukin 10 (IL-10) inhibits cytokine synthesis by human monocytes: an autoregulatory role of IL-10 produced by monocytes.
J Exp Med
174
1991
1209
155
Wang
P
Wu
P
Siegel
MI
Egan
RW
Billah
MM
IL-10 inhibits transcription of cytokine genes in human peripheral blood mononuclear cells.
J Immunol
153
1994
811
156
Cox
RA
Anders
GT
Cappelli
PJ
Johnson
JE
Blanton
HM
Seaworth
BJ
Treasure
RL
Production of tumor necrosis factor-alpha and interleukin-1 by alveolar macrophages from HIV-1-infected persons.
AIDS Res Hum Retroviruses
6
1990
431
157
Navikas
V
Link
J
Wahren
B
Persson
C
Link
H
Increased levels of interferon-gamma (IFN-gamma), IL-4 and transforming growth factor-beta (TGF-beta) mRNA expressing blood mononuclear cells in human HIV infection.
Clin Exp Immunol
96
1994
59
158
Rossol-Voth
R
Klein
SA
Geissler
G
Gute
P
Helm
EB
Falke
D
Hoelzer
D
TNF-alpha produced by gamma delta-T-cells as the pivotal pathogenic factor in HIV-infection for the depletion of CD4-pos. T-cells and the inhibition of the hematopoiesis.
Int Conf AIDS
8
1992
A69
159
Husain
SR
Leland
P
Aggarwal
BB
Puri
RK
Transcriptional up-regulation of interleukin 4 receptors by human immunodeficiency virus type 1 tat gene.
AIDS Res Hum Retroviruses
12
1996
1349
160
Brady
HJ
Abraham
DJ
Pennington
DJ
Miles
CG
Jenkins
S
Dzierzak
EA
Altered cytokine expression in T lymphocytes from human immunodeficiency virus Tat transgenic mice.
J Virol
69
1995
7622
161
Gallicchio
VS
Tse
KF
Morrow
J
Hughes
NK
Suppression of hematopoietic support function is associated with overexpression of interleukin-4 and transforming growth factor-b1 in LP-BM5 murine-leukemia-virus-infected stromal cell lines.
Acta Haematol
95
1996
204
162
Gautam
SC
Noth
CJ
Janakiraman
N
Pindolia
KR
Chapman
RA
Induction of chemokine mRNA in bone marrow stromal cells: Modulation by TGF-beta 1 and IL-4.
Exp Hematol
23
1995
482
163
Sunila
I
Vaccarezza
M
Pantaleo
G
Fauci
AS
Orenstein
JM
Gp120 is present on the plasma membrane of apoptotic CD4 cells prepared from lymph nodes of HIV-1-infected individuals: An immunoelectron microscopic study.
AIDS
11
1997
27
164
Kolesnitchenko
V
Wahl
LM
Tian
H
Sunila
I
Tani
Y
Hartmann
DP
Cossman
J
Raffeld
M
Orenstein
J
Samelson
LE
Human immunodeficiency virus 1 envelope-initiated G2-phase programmed cell death.
Proc Natl Acad Sci USA
92
1995
11889
165
Takenaka
K
Nagafuji
K
Harada
M
Miyamoto
T
Makino
S
Gondo
H
Okamura
T
Niho
Y
In vitro expansion of hematopoietic progenitor cells induces functional expression of fas antigen (CD95).
Blood
88
1996
2871
166
Barcena
A
Park
SW
Banapour
B
Muench
MO
Mechetner
E
Expression of Fas/CD95 and Bcl-2 by primitive hematopoietic progenitors freshly isolated from human fetal liver.
Blood
88
1996
2013
167
Maciejewski
JP
Weichold
FF
Young
NS
HIV-1 suppression of hematopoiesis in vitro mediated by envelope glycoprotein and TNF-alpha.
J Immunol
153
1994
4303
168
Roux-Lombard
P
Modoux
C
Cruchaud
A
Dayer
J-M
Purified blood monocytes from HIV 1-infected patients produce high levels of TNFa and IL-1.
Clin Immunol Immunopathol
50
1989
374
169
Zauli
G
Davis
BR
Re
MC
Visani
G
Furlini
G
La Placa
M
tat Protein stimulates production of transforming growth factor-b1 by marrow macrophages: A potential mechanism for human immunodeficiency virus-1-induced hematopoietic suppression.
Blood
80
1992
3036
170
Lotz
M
Seth
P
TGF beta and HIV infection.
Ann NY Acad Sci
685
1993
501
171
Denis
M
Ghadirian
E
Alveolar macrophages from subjects infected with HIV-1 express macrophage inflammatory protein-1a (MIP-1a): Contribution to the CD8+ alveolitis.
Clin Exp Immunol
96
1994
187
172
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 beta 1.
J Exp Med
183
1996
99
173
Folks
TM
Kessler
SW
Orenstein
JM
Justement
JS
Jaffe
ES
Fauci
AS
Infection and replication of HIV-1 in purified progenitor cells of normal human bone marrow.
Science
242
1988
919
174
Steinberg
HN
Crumpacker
CS
Chatis
PA
In vitro suppression of normal human bone marrow progenitor cells by human immunodeficiency virus.
J Virol
65
1991
1765
175
Kojouharoff G, Ottmann OG, Von Briesen H, Geissler G, R:ubsamen-Waigmann H, Hoelzer D, Ganser A: Infection of granulocyte/monocyte progenitor cells with HIV1. Res Virol 142:151, 1991
176
Zauli
G
Re
MC
Giovannini
M
Bagnara
GP
Furlini
G
Ranieri
S
La Placa
M
Effect of human immunodeficiency virus type 1 on CD34+ cells.
Ann NY Acad Sci
628
1991
273
177
Zauli
G
Re
MC
Visani
G
Furlini
G
La Placa
M
Inhibitory effect of HIV-1 envelope glycoproteins gp120 and gp160 on the in vitro growth of enriched (CD34+) hematopoietic progenitor cells.
Arch Virol
122
1992
271
178
Calenda
V
Sebahoun
G
Chermann
JC
Modulation of normal human erythropoietic progenitor cells in long-term liquid cultures after HIV-1 infection.
AIDS Res Hum Retroviruses
8
1992
61
179
Cen
D
Zauli
G
Szarnicki
R
Davis
BR
Effect of different human immunodeficiency virus type-1 (HIV-1) isolates on long-term bone marrow haemopoiesis Effect of different human immunodeficiency virus type-1 (HIV-1) isolates on long-term bone marrow haemopoiesis.
Br J Haematol
85
1993
596
180
Schwartz
GN
Kessler
SW
Szabo
JM
Burrell
LM
Francis
ML
Negative regulators may mediate some of the inhibitory effects of HIV-1 infected stromal cell layers on erythropoiesis and myelopoiesis in human bone marrow long term cultures.
J Leukoc Biol
57
1995
948
181
Kaushal
S
La Russa
VF
Gartner
S
Kessler
S
Perfetto
S
Yu
Z
Ritchey
DW
Xu
J
Perera
P
Kim
J
Reid
T
Mayers
DL
St. Louis
D
Mosca
JD
Exposure of human CD34+ cells to human immunodeficiency virus type 1 does not influence their expansion and proliferation of hematopoietic progenitors in vitro.
Blood
88
1996
130
182
Donahue
RE
Johnson
MM
Zon
LI
Clark
SC
Groopman
JE
Suppression of in vitro haematopoiesis following human immunodeficiency virus infection.
Nature
326
1987
200
183
Von Laer
D
Hufert
FT
Fenner
TE
Schwander
S
Dietrich
M
Schmitz
H
Kern
P
CD34+ hematopoietic progenitor cells are not a major reservoir of the human immunodeficiency virus.
Blood
76
1990
1281
184
Ganser
A
Ottmann
OG
Von Briesen
H
Volkers
B
Rubsamen-Waigmann
H
Hoelzer
D
Changes in the haematopoietic progenitor cell compartment in the acquired immunodeficiency syndrome.
Res Virol
141
1990
185
185
Zauli
G
Re
MC
Davis
B
Sen
L
Visani
G
Gugliotta
L
Furlini
G
La Placa
M
Impaired in vitro growth of purified (CD34+) hematopoietic progenitors in human immunodeficiency virus-1 seropositive thrombocytopenic individuals.
Blood
79
1992
2680
186
Louache
F
Henri
A
Bettaieb
A
Oksenhendler
E
Raguin
G
Tulliez
M
Vainchenker
W
Role of human immunodeficiency virus replication in defective in vitro growth of hematopoietic progenitors.
Blood
80
1992
2991
187
Kaczmarski
RS
Davison
F
Blair
E
Sutherland
S
Moxham
J
McManus
T
Mufti
GJ
Detection of HIV in haemopoietic progenitors.
Br J Haematol
82
1992
764
188
Dallalio
G
North
M
Means
RT
Jr
Inhibition of marrow CFU-E colony formation from human immunodeficiency virus-infected patients by b- and gamma-interferon.
Am J Hematol
53
1996
118
189
Marandin
A
Katz
A
Oksenhendler
E
Tulliez
M
Picard
F
Vainchenker
W
Louache
F
Loss of primitive hematopoietic progenitors in patients with human immunodeficiency virus infection.
Blood
88
1996
4568
Sign in via your Institution