The human T-cell leukemia virus type I (HTLV-I) regulatory protein, Tax, has been speculated to play a major role in HTLV-I leukemogenesis. Indeed, several studies have suggested that upregulation of various cellular oncogenes and cytokines by Tax may explain the pathogenesis observed in HTLV-I–infected individuals, as well as several Tax-transgenic animal models. We report here the analysis of cytokine expression in a Tax-transgenic animal model with large granular lymphocytic (LGL) leukemia. Two different transgenic mice showed identical expression of interleukin-1α (IL-1α), IL-1β, interferon γ (IFNγ), and granulocyte-macrophage colony-stimulating factor (GM-CSF ) in peripheral tail tumors. Interestingly, LGL cell lines derived from these same tumors expressed high levels of both IFNγ and GM-CSF, which correlated with the level of Tax expression. These same LGL cell lines also expressed high levels of lymphocyte function-associated antigen-1 (LFA-1) and intracellular adhesion molecule-1 (ICAM-1). Engraftment of these LGL cell lines into severe combined immunodeficient (SCID) mice led to the development of leukemia and lymphomas. Examination of these SCID mice showed that their pathology was nearly identical to that observed in the original Tax-transgenic mouse model. Both the Tax-transgenic and engrafted SCID mouse models allow for the analysis of cellular events that are required for tumor development associated with HTLV infection and suggest that Tax expression may be responsible for the upregulation of certain cytokines and adhesion molecules that affect the infiltrating capabilities of HTLV-I–infected cells.

HUMAN T-CELL leukemia virus type I (HTLV-I) was the first pathogenic retrovirus discovered in humans and is the etiologic agent of adult T-cell leukemia/lymphoma (ATLL) and tropical spastic paraparesis (TSP).1,2 HTLV infection has also been associated with other clinical disorders such as arthropathy,3 uveitis,4 mycosis fungoides,5 and large granular lymphocytic (LGL) leukemia.6,7 The mode of leukemic transformation by HTLV infection is still unclear, but has been suggested to involve the HTLV regulatory protein, Tax, which has been shown to be a potent transcriptional activator of the HTLV long terminal repeat as well as several cellular genes.8-14 It is this promiscuous transactivating activity of Tax that has been postulated to be the mode of HTLV transformation. Indeed, expression of Tax alone has been shown to transform a number of cell types in vitro, including 3T3, Rat-1,15 Rat-2,16 primary rat embryo fibroblasts,17 and primary human CD4+ lymphocytes.18 

The development of animal model systems to study the cellular events that are required for in vivo leukemogenesis and tumor transformation by HTLV is crucial.19 Several transgenic mouse models have been developed that express HTLV-I–Tax under a variety of enhancers/promoters.20-24 Although many of these transgenic mice developed neoplasias of nonlymphoid compartments, none of them resulted in the development of lymphomas and/or leukemia as a result of Tax expression. Previously, we have reported the generation of transgenic mice that target expression of Tax to the mature T-lymphocyte/natural killer (NK)-cell compartment by using the human granzyme B (GzmB) promoter.25 These mice developed LGL leukemia/lymphoma of T/NK-cell origin, showing that Tax expression in the lymphoid compartment is sufficient for the induction of leukemia. From this transgenic model, we were able to establish several T/NK-like LGL cell lines that were morphologically and phenotypically identical to those observed in vivo. We report here the differential expression of cytokines expressed in vivo and from established LGL cell lines, which may help to elucidate the cellular events that lead to the progression of tumor development in this transgenic model. In addition, we show that these LGL cell lines are capable of being engrafted into the severe combined immunodeficiency (SCID) mouse and transferring a nearly identical pathogenic phenotype as that observed in the original GzmB-Tax transgenic mice.

Mice.Immunodeficient SCID (Balb/C.B17 scid/scid) mice were a gift from H. Skip Virgin and Samuel Stanley (Washington University, St Louis, MO). The GzmB-Tax transgenic mice were generated using the 5′ flanking region (−1170 to +36) of the human granzyme B gene as previously described.25 All mice were bred and maintained under pathogen-free conditions in accordance with Washington University School of Medicine animal care guidelines.

Cells.Tumor cell suspensions were prepared from founder F8 and F1 progeny (m388, f197) of founder M9 using sterilized frosted glass slides and cultured in Iscove's minimal essential medium with 10% (vol/vol) fetal calf serum (GIBCO, Grand Island, NY), 1% L-glutamine, 1% penicillin/streptomycin, and interleukin-2 (IL-2) at 500 to 1,000 U/mL (Cetus, Sunnyvale, CA). Cell suspensions were propagated for greater than 2 years in vitro, from which the homogeneous LGL cell lines (F8, m388, and f197) were derived. The SC cell line was derived from a peritoneal tumor of a SCID mouse engrafted with the F8 LGL cell line.

Injection of the cells into SCID mice.F8 cells (1 to 10 × 107) were washed two times in Iscove's medium. These cells were then resuspended in 1 mL of Iscove's medium and were intraperitoneally injected into SCID mice (F8 LGL cell line-injected). Mock-injected SCID mice were injected intraperitoneally with Iscove's medium.

Tissue histology and peripheral blood analysis.Tissues were fixed in 10% neutral-buffered formalin (Sigma, St Louis, MO), embedded in paraffin (Baxter, Deerfield, IL) for serial sectioning, and stained with hematoxylin/eosin (H&E; Sigma). Peripheral blood was analyzed on a Baker 9000 Coulter Counter (Serono Laboratories, Randolph, MA) and differential cell counts were reported as the mean cell count per 100 cells examined microscopically. Wright-Giemsa (Fisher, Pittsburgh, PA) staining was performed on peripheral blood smears.

Fig. 1.

Flow cytometric analysis of cell surface markers on Tax-transgenic LGL cell lines. Reactivity of antibodies against cell surface markers is indicated with solid lines. Reactivity of a control antibody, indicated by dashed lines, is depicted in all panels.

Fig. 1.

Flow cytometric analysis of cell surface markers on Tax-transgenic LGL cell lines. Reactivity of antibodies against cell surface markers is indicated with solid lines. Reactivity of a control antibody, indicated by dashed lines, is depicted in all panels.

Close modal

Reverse transcription-polymerase chain reaction (RT-PCR) for cytokine analysis.RNA from crude tumor cell suspensions and LGL cell lines was purified with guanidinium isothiocyanate as previously described.25 Eppendorfs containing 10 μg of total RNA were heated for 10 minutes at 65°C and chilled on ice for 5 minutes. Each sample was then incubated for 40 minutes at 37°C after adding 1 μL RNasin (Promega, Madison, WI), 2 μg oligo dT(12-18) (GIBCO-BRL, Gaithersburg, MD), 0.8 mmol/L deoxynucleoside triphosphates (US Biochemicals [USB], Cleveland, OH), 400 U reverse transcriptase (Moloney's murine leukemia virus [M-MLV]; USB), 10 μL of 5× reverse transcriptase buffer (USB), and diethyl procarbonate (DEPC)-treated sterile water to adjust the volume to 50 μL. An additional 400 U of reverse transcriptase was added to the reaction and the samples were incubated for an additional 40 minutes before heat-inactivating the reaction at 65°C for 10 minutes. The total volume was then adjusted to 100 μL and stored at −20°C until use.

PCR.Ten microliters of each RT-PCR sample was added to a 100 μL reaction mixture (1× PCR buffer, 0.2 mmol/L dNTPs, 2.5 U Taq DNA polymerase, 25 mmol/L MgCl2 , 0.5 μg each of 3′ and 5′ primers; USB). All reaction tubes included a 50-μL mineral oil overlay. The PCR was run in a DNA Thermal Cycler (Perkin Elmer, Norwalk, CT) as follows: 1 minute of hot start at 93°C; 30 cycles of 93°C for 1 minute, 60°C for 2 minutes, and 72°C for 1 minute; and 3 minutes of final extension at 72°C. In all experiments, negative controls were performed whereby RNA samples, which were not reverse transcribed, were amplified with primers for the murine housekeeping gene hypoxanthine phosphoribosyltransferase (HPGRT). These negative controls were performed in parallel under identical conditions as experimental samples and are indicated as HPGRT(−) samples. All cytokine primers were designed to span introns, thus ensuring that they would not detect genomic DNA sequences. Cytokine primers for IL-1α, IL-1β, IL-2, IL-4, and interferon γ (IFNγ) were a gift from Paul Allen (Washington University) and were designed at DNAX Research Institute (Palo Alto, CA). Cytokine primers for granulocyte-macrophage colony-stimulating factor (GM-CSF ) have been previously described.26 Cytokine primers for the internal control gene HPGRT were designed by Kenneth Murphy (Washington University). The following are sequences of the cytokine amplification primers (S, sense; A, antisense) with their predicted product size in parentheses: IL-1α, S 5′-CTCTAGAGCACCATGCTACAGAC-3′ (308); IL-1α A, 5′-TGGAATCCAGGGGAAACACTG-3′; IL-1β S, 5′-TGAAGGGCTGCTTCCAAACCTTTGACC-3′ (322); IL-1β A, 5′-TGTCCATTGAGGTGGAGAGCTTTCAGC-3′; IL-2 S, 5′-TGATGGACCTACAGGAGCTCCTGAG-3′ (167); IL-2 A, 5′-GAGTCAAATCCAGAACATGCCGCAG-3′; IL-4 S, 5′ACGAGGTCACAGGAGAAGGGACGCCATGCA-3′ (188); IL-4 A, 5′-TCATTCATGGAGCAGCTTATCGATCGAATCC-3′; IFNγ S, 5′-TGGAGGAACTGGCAAAAGGATGGT-3′ (336); IFNγ A, 5′-TTGGGACAATCTCTTCCCCAC-3′; GM-CSF S, 5′-TTCCTGGGCATTGTGGTCT-3′ (430); GM-CSF A, 5′-TGGATTCAGAGCTGGCCTGG-3′; HPGRT S, 5′-TGGGAGGCCATCACATTGT-3′ (471); HPGRT A, 5′-GCTTTTCCAGTTTCACTAATGACA-3′.

PCR samples were separated by electrophoresis in a 2% agarose gel (GIBCO-BRL) in Tris borate/EDTA buffer containing 0.5 μg/mL ethidium bromide. Bands were visualized by UV light exposure and photographed using a Stratagene EagleEye II (Stratagene, La Jolla, CA).

Enzyme-linked immunosorbent assay (ELISA) measurement of cytokines.Supernatant was obtained from 1 × 107 cells seeded in 10 mL of complete Iscove's medium and quantitated for cytokine production using the two-antibody sandwich assay. The monoclonal antibody H22, used as the capture antibody against murine IFNγ, was a gift from Robert Schreiber (Washington University). The secondary goat antimouse IFNγ antibody and recombinant murine IFNγ were gifts from Kenneth Murphy (Washington University). Detection in the IFNγ ELISA was performed using a peroxidase-conjugated rabbit antigoat tertiary antibody (Sigma) and the chromogenic substrate 2,2′-azino-bis(3-ethylbenzthiazoline-6)-sulfonic acid (Sigma). Substrate conversion was measured by a spectrophotometer at 405 nm. GM-CSF cytokine production was quantitated using the mouse GM-CSF Quantikine M Immunoassay (R & D Systems, Rochester, MN). All samples were performed in triplicate.

Fig. 2.

RT-PCR cytokine analysis of GzmB-Tax transgenic tumors and derived LGL cell lines. The first and third panels show mRNA expression of IL-1α, IL-1β, IFNγ, and GM-CSF in the peripheral tumors of two different transgenic mice (m388 Tumor and f197 Tumor). The second and fourth panels show that the tumor-derived LGL cell lines express mRNAs for only IFNγ and GM-CSF (m388 LGL Cell Line and f197 LGL Cell Line). Negative and positive RT-PCR controls are shown in each panel and are labeled as HPGRT(−) and HPGRT(+), respectively. Excess primer for each RT-PCR reaction is indicated at the right side of each panel (primer).

Fig. 2.

RT-PCR cytokine analysis of GzmB-Tax transgenic tumors and derived LGL cell lines. The first and third panels show mRNA expression of IL-1α, IL-1β, IFNγ, and GM-CSF in the peripheral tumors of two different transgenic mice (m388 Tumor and f197 Tumor). The second and fourth panels show that the tumor-derived LGL cell lines express mRNAs for only IFNγ and GM-CSF (m388 LGL Cell Line and f197 LGL Cell Line). Negative and positive RT-PCR controls are shown in each panel and are labeled as HPGRT(−) and HPGRT(+), respectively. Excess primer for each RT-PCR reaction is indicated at the right side of each panel (primer).

Close modal

Flow cytometry and Western blot analysis.Tumor cell lines (1 × 106 cells) were incubated with 5 μg of unlabeled Fcγ receptor (FcγRII/III) antibody for 15 minutes at 4°C to block free surface FcγR (except when staining for this particular surface marker) and then counterstained for 30 minutes at 4°C with fluorescein isothiocyanate– or R-phycoerythrin–conjugated antibodies to the following surface markers: CD3ε, CD4, CD8β chain, TCR α/β, CD90.2 (Thy1.2), CD122 (IL-2Rβ chain), CD25 (IL-2Rα chain), H2Kk, PK136 (NK1.1), CD49d (VLA-4), CD11a (lymphocyte function-associated antigen-1 [LFA-1] α chain), CD54 (intracellular adhesion molecule-1 [ICAM-1]), and CD11b (Mac-1) from PharMingen (San Diego, CA); and 5E6 (Ly49C) and A1 (Ly49A) from W. Yokoyama (Washington University). These cell lines were then analyzed on a FACScan (Becton Dickinson, Mountain View, CA). For Western blot analysis, 107 cells from the F8 cell line, SC cell line, f197 cell line, m388 cell line, MT2 HTLV-I–positive cell line, and the Jurkat E6.1 HTLV-I–negative cell line were lysed in 200 μL of 2× sample buffer (1 mmol/L EDTA, 0.125 mol/L Tris [pH 6.8], 4% sodium dodecyl sulfate [SDS], 10% β-mercapatoethanol [β-ME], 30% glycerol, 0.01% bromophenol blue). These samples were heated at 85°C for 10 minutes and centrifuged at 14,000 rpm to clarify, and then 10 to 30 μL of each sample was subjected to a 10% SDS-polyacrylamide gel electrophoresis (SDS-PAGE) gel. Gels were transferred to nitrocellulose, probed (1:5,000) with a pool of purified anti-Tax monoclonal antibodies (nos. 168A51-2, 168A51-42, and 168B17-46-34; AIDS Reagent Program, Rockville, MD),27 and visualized by using enhanced chemiluminescence (Amersham, Arlington Heights, IL).

Cell proliferation assay.The MTT proliferation assay was essentially performed according to Hansen et al.28 Briefly, 50,000 cells from each cell line were plated in triplicate into flat-bottom 96-well plates in 100 μL of complete Iscove's medium. To each of set of triplicates was added 100 μL of media or media containing cytokines to give the indicated final concentrations: IL-2 (500 U/mL; Cetus), GM-CSF (20 ng/mL), IL-1α and IL-1β (25 ng/mL), and IFNγ (250 U/mL; R & D Systems). These cultures were incubated at 37°C for 3 days, at which time 25 μL of MTT [(3,(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide) 5 mg/mL in phosphate-buffered saline (Sigma)] was added to each well and allowed to incubate at 37°C for an additional 4 hours. Each sample was lysed (20% wt/vol of SDS/50% DMF in dH2O, pH 4.7) overnight at 37°C, and their optical densities at 562 nm were measured.

Fig. 3.

MTT cell proliferation assay on LGL cell lines. (A) LGL cell lines were incubated with or without IL-2 and measured for their proliferation. As shown, the F8 and SC LGL cell lines have become independent of IL-2 for proliferation, but have retained their capability to respond to IL-2 (*P < .05). In contrast, the f197 and m388 cell lines are highly dependent on exogenous IL-2 for their proliferation (**P < .01). (B) The IL-2–dependent LGL cell lines, f197 and m388, were incubated with the cytokines found to be expressed in the peripheral tumor from which they were derived (IL-1α and IL-1β, IFNγ, and GM-CSF ) and examined for their proliferative effects. Incubation with IL-2 and media alone served as positive and negative controls, respectively. As shown, none of these cytokines was able to support the proliferation of these IL-2–dependent LGL cell lines.

Fig. 3.

MTT cell proliferation assay on LGL cell lines. (A) LGL cell lines were incubated with or without IL-2 and measured for their proliferation. As shown, the F8 and SC LGL cell lines have become independent of IL-2 for proliferation, but have retained their capability to respond to IL-2 (*P < .05). In contrast, the f197 and m388 cell lines are highly dependent on exogenous IL-2 for their proliferation (**P < .01). (B) The IL-2–dependent LGL cell lines, f197 and m388, were incubated with the cytokines found to be expressed in the peripheral tumor from which they were derived (IL-1α and IL-1β, IFNγ, and GM-CSF ) and examined for their proliferative effects. Incubation with IL-2 and media alone served as positive and negative controls, respectively. As shown, none of these cytokines was able to support the proliferation of these IL-2–dependent LGL cell lines.

Close modal

Phenotypic analysis of immortalized Tax-transgenic LGL cell lines.Three separate T/NK-like LGL cell lines were established from the original GzmB-Tax transgenic mice.25 These cell lines were derived from the peripheral tumors of three different mice of two different founder lines. One LGL cell line (F8) exhibited cell surface markers consistent with that of an NK-cell lineage, namely expression of FcγRII/III and the IL-2Rβ chain (Fig 1). Some of these cells did express low levels of TCR α/β and CD3ε (22% and 11%, respectively); however, these cells were a minority of the bulk population. The F8 cell line did not express any of the classical NK-cell markers, namely NK1.1, Ly49A, or Ly49C (data not shown). The other two LGL cell lines (f197 and m388) exhibited cell surface markers of the T-cell lineage, specifically CD3ε, TCR α/β, Thy1.2, and the IL-2Rβ chain. Interestingly, both of these cell lines also expressed the IL-2Rα chain (CD25), which is a characteristic surface marker of most HTLV-I–infected cells.29,30 None of the cell lines expressed CD4 (data not shown); however, the f197 cell line expressed CD8, a marker that is found on HTLV-II–infected cells and only occasionally found on HTLV-I–infected cells (Fig 1).31 This heterogeneity of distinct immortalized cell phenotypes from the tumors of different mice is consistent with the activity of the granzyme B promoter32 as well as the multiple-hit theory needed for tumor development.

Fig. 4.

ELISAs for GM-CSF and IFNγ production of tumor-derived LGL cell lines. (A) GM-CSF ELISA showing production of soluble GM-CSF by all four cell lines (f197, 867± 75 pg/mL; m388, 620± 56 pg/mL; F8, 127 ± 9 pg/mL; and SC, 141 ± 9 pg/mL). The production of GM-CSF between IL-2–dependent cell lines (f197 and m388) is significantly different from that of the IL-2–independent cell lines (F8 and SC; **P < .01). (B) IFNγ ELISA showing production of soluble IFNγ by all four cell lines (f197, 288 ± 18 U/mL; m388, 212 ± 15 U/mL; F8, 119 ± 12 U/mL; and SC, 108 ± 11 U/mL). Similarly, the production of IFNγ between IL-2–dependent cell lines and IL-2–independent cell lines is significantly different (**P < .01).

Fig. 4.

ELISAs for GM-CSF and IFNγ production of tumor-derived LGL cell lines. (A) GM-CSF ELISA showing production of soluble GM-CSF by all four cell lines (f197, 867± 75 pg/mL; m388, 620± 56 pg/mL; F8, 127 ± 9 pg/mL; and SC, 141 ± 9 pg/mL). The production of GM-CSF between IL-2–dependent cell lines (f197 and m388) is significantly different from that of the IL-2–independent cell lines (F8 and SC; **P < .01). (B) IFNγ ELISA showing production of soluble IFNγ by all four cell lines (f197, 288 ± 18 U/mL; m388, 212 ± 15 U/mL; F8, 119 ± 12 U/mL; and SC, 108 ± 11 U/mL). Similarly, the production of IFNγ between IL-2–dependent cell lines and IL-2–independent cell lines is significantly different (**P < .01).

Close modal
Fig. 5.

Immunoblot analysis of Tax expression in LGL cell lines. Tax expression is indicated by the arrow on the left, with molecular mass markers shown on the right. LGL cell line dependency for IL-2 is indicated below each sample lane. Jurkat and MT2 cells were used for negative and positive controls of Tax expression, respectively.

Fig. 5.

Immunoblot analysis of Tax expression in LGL cell lines. Tax expression is indicated by the arrow on the left, with molecular mass markers shown on the right. LGL cell line dependency for IL-2 is indicated below each sample lane. Jurkat and MT2 cells were used for negative and positive controls of Tax expression, respectively.

Close modal

Cytokine mRNA expression in tumors and derived cell lines.Tax has been shown to upregulate the expression of numerous cytokines,13,14,33-37 some of which have been speculated to play a role in the pathogenesis observed in ATLL and TSP. Because of these observations, we decided to examine the differential mRNA expression of IL-1α, IL-1β, IL-2, IL-4, IFNγ, and GM-CSF for their possible contribution to the pathogenesis/tumor formation observed in the GzmB-Tax mice. RT-PCR was performed on RNA obtained from the peripheral tail tumors of two different GzmB-Tax transgenic mice (m388 and f197) as well as RNA from LGL cell lines derived from these tumors. Both mice exhibited identical cytokine profiles from their tumors, which were shown to express IL-1α, IL-1β, IFNγ, and GM-CSF (Fig 2). Interestingly, both LGL cell lines also displayed identical cytokine profiles. These derived cell lines were shown to express both IFNγ and GM-CSF, suggesting that the LGL cells may be responsible for the IFNγ and GM-CSF expression observed in the original tumor (Fig 2). Because T cells and NK cells have been shown to produce both IFNγ and GM-CSF,38-40 expression of these cytokines by the m388 and f197 LGL cell lines gives additional support that these cells are of T/NK-cell origin.

Table 1.

LGL Cell Line Tumorigenicity in SCID Mice

Cell LineIL-2 Dependent (+ or −)Cells Injected (×10−7)Tumor Growth/No. of MiceSurvival Time (mo)Organ Involvement
LNLungSpleen*PBLiver
F8 − 10 2/2 2.5 ++ ++ ++ 
F8 − 2/2 ++ ++ ++ 
F8 − 1/1 2.5 ++ ++ ND 
F8 − 1/1 ++ ++ ++ +/− 
f197 10 1/4 >6 − − 
m388 10 0/4 >6 − − − − − 
Cell LineIL-2 Dependent (+ or −)Cells Injected (×10−7)Tumor Growth/No. of MiceSurvival Time (mo)Organ Involvement
LNLungSpleen*PBLiver
F8 − 10 2/2 2.5 ++ ++ ++ 
F8 − 2/2 ++ ++ ++ 
F8 − 1/1 2.5 ++ ++ ND 
F8 − 1/1 ++ ++ ++ +/− 
f197 10 1/4 >6 − − 
m388 10 0/4 >6 − − − − − 

Abbreviations: ND, not determined; LN, lymph node; PB, peripheral blood (LGL lymphocytosis); ++, marked infiltration; +, slight infiltration.

*

Structural integrity compromised.

Fig. 6.

Pathohistological features of the tissues of SCID mice engrafted with the transformed Tax-transgenic F8 LGL cell line. (A) Section of a peritoneal tumor located at the site of injection, showing that it is comprised primarily of LGLs (original magnification × 720). (B) Section of a mesenteric lymph node showing massive LGL infiltration, characteristic of diffuse large-cell type lymphoma (original magnification × 1,080). (C) Section of a spleen showing infiltration of LGLs in the left portion with generalized disruption of both the white and red pulp (original magnification × 720). (D) Section of a liver showing infiltration of LGLs into the sinusoid and portal region (open arrows; original magnification × 360). (E) Section of a mock-injected SCID lung showing an intrapulmonary bronchus with associated epithelium and normal alveolar ducts and alveoli (original magnification × 360). (F ) Lung section from an LGL-engrafted SCID mouse showing massive infiltration of LGLs into the bronchus epithelium and alveoli septa (original magnification × 360). (G) Peripheral blood smear from a mock-injected SCID mouse showing a paucity of circulating lymphocytes and neutrophils (original magnification × 360). (H) Peripheral blood smear from an LGL-engrafted SCID mouse demonstrating LGL lymphocytosis and neutrophilia (original magnification × 720). (I) Higher magnification of (H) showing morphologic characteristics of the LGLs (original magnification × 2,100).

Fig. 6.

Pathohistological features of the tissues of SCID mice engrafted with the transformed Tax-transgenic F8 LGL cell line. (A) Section of a peritoneal tumor located at the site of injection, showing that it is comprised primarily of LGLs (original magnification × 720). (B) Section of a mesenteric lymph node showing massive LGL infiltration, characteristic of diffuse large-cell type lymphoma (original magnification × 1,080). (C) Section of a spleen showing infiltration of LGLs in the left portion with generalized disruption of both the white and red pulp (original magnification × 720). (D) Section of a liver showing infiltration of LGLs into the sinusoid and portal region (open arrows; original magnification × 360). (E) Section of a mock-injected SCID lung showing an intrapulmonary bronchus with associated epithelium and normal alveolar ducts and alveoli (original magnification × 360). (F ) Lung section from an LGL-engrafted SCID mouse showing massive infiltration of LGLs into the bronchus epithelium and alveoli septa (original magnification × 360). (G) Peripheral blood smear from a mock-injected SCID mouse showing a paucity of circulating lymphocytes and neutrophils (original magnification × 360). (H) Peripheral blood smear from an LGL-engrafted SCID mouse demonstrating LGL lymphocytosis and neutrophilia (original magnification × 720). (I) Higher magnification of (H) showing morphologic characteristics of the LGLs (original magnification × 2,100).

Close modal

Cytokine responsiveness of LGL cell lines.All three LGL cell lines exhibited an initial growth dependence for high IL-2 concentrations. Two of these cell lines have retained this dependency for more than 2 years (m388 and f197), whereas one cell line has gained IL-2 independence (F8), but has retained the capability to respond to IL-2 (Fig 3A; P < .05). To examine whether any of the other cytokines detected in these peripheral tumors (IL-1α, IL-1β, IFNγ, and GM-CSF ) may play a role in the proliferation of the IL-2–dependent LGL cell lines (f197 and m388), a proliferation assay was performed that showed that none of these cytokines were able to support the proliferation of these cells either alone or in any combination (Fig 3B, data not shown).

Soluble IFNγ and GM-CSF levels produced by LGL cells and their relationship to Tax protein levels.To ensure that the m388 and f197 LGL cell lines produced soluble IFNγ and GM-CSF, ELISAs were performed on the supernatant from these cultured cell lines. In addition, two other IL-2–independent LGL cell lines (F8 and SC) were similarly tested. All four cell lines produced substantial levels of both cytokines. Interestingly, the IL-2–dependent cell lines (m388 and f197) produced significantly more IFNγ and GM-CSF than the IL-2–independent cell lines (F8 and SC; Fig 4A and B; P < .001). One possible explanation for the higher cytokine production in the IL-2–dependent LGL cell lines could be due to higher levels of Tax expression in these cell lines compared with the IL-2–independent cell lines. Immunoblot analysis showed significant levels of Tax protein in the IL-2–dependent cell lines, but nearly undetectable levels in the IL-2–independent cell lines (Fig 5). The lack of Tax expression in the IL-2–independent cell lines was not due to loss of transgene because Southern blot analysis of all cell lines showed identical integration patterns as originally described (data not shown). These results show a plausible correlation between high cytokine expression and high levels of Tax expression, compatible with previous observations of Tax's ability to upregulate the expression of both IFNγ and GM-CSF.14 41 

Table 2.

Peripheral Blood Analysis of Engrafted LGL-SCID Mice

Total and Differential WBC Counts in Tumor-Injected and Mock-Injected SCID Mice
Mouse IDWBCRBC% of Nucleated CellsHbR%
LNMELGL
F8 LGL cell line-injected 
SC-1 10.5 4.85 11 58 30 7.5 18.2 
SC-2 10.7 4.80 72 21 7.3 9.8 
SC-3 9.7 4.63 71 22 7.2 12.7 
SC-4 13.5 6.63 70 20 9.0 2.4 
Average 11.1* 5.22* 7* 68 23* 7.8* 10.8Mock-injected 
NML-1 2.0 10.22 59 40 14.8 0.7 
NML-2 1.8 8.94 53 45 13.3 2.9 
NML-3 1.6 8.62 49 48 12.9 1.3 
NML-4 4.5 9.44 40 58 13.7 0.8 
NML-5 3.7 9.21 42 54 13.8 2.0 
Average 2.7 9.29 49 49 13.7 1.5 
Total and Differential WBC Counts in Tumor-Injected and Mock-Injected SCID Mice
Mouse IDWBCRBC% of Nucleated CellsHbR%
LNMELGL
F8 LGL cell line-injected 
SC-1 10.5 4.85 11 58 30 7.5 18.2 
SC-2 10.7 4.80 72 21 7.3 9.8 
SC-3 9.7 4.63 71 22 7.2 12.7 
SC-4 13.5 6.63 70 20 9.0 2.4 
Average 11.1* 5.22* 7* 68 23* 7.8* 10.8Mock-injected 
NML-1 2.0 10.22 59 40 14.8 0.7 
NML-2 1.8 8.94 53 45 13.3 2.9 
NML-3 1.6 8.62 49 48 12.9 1.3 
NML-4 4.5 9.44 40 58 13.7 0.8 
NML-5 3.7 9.21 42 54 13.8 2.0 
Average 2.7 9.29 49 49 13.7 1.5 

Abbreviations: Total WBC count is expressed as number (×10−12) per liter; Hb, hemoglobin expressed as g/dL; L, lymphocytes; N, neutrophils; M, monocytes; E, eosinophils; R, reticulocytes; LGL, large granular lymphocytes; RBC, red blood cells.

*

Significantly different from mock-injected SCID mice (P < .001).

Significantly different from mock-injected SCID mice (P < .01).

Significantly different from mock-injected SCID mice (P < .05).

Engraftment of immortalized LGL cells in SCID mice.The immortalized F8 LGL cell line was subsequently examined for its state of transformation by examining its ability to engraft in SCID mice. Six SCID mice were inoculated intraperitoneally with 1 to 10 × 107 F8 cells. All six F8-injected SCID mice developed LGL leukemia and lymphomas, resulting in the death of the SCID mice after 2 to 2.5 months (Table 1). Examination of the F8-injected SCID mice showed a large tumor mass in the peritoneal cavity, composed primarily of LGLs (Fig 6A). In addition, LGL infiltration was observed in numerous organs, including lymph nodes, spleen, liver, lung, and peripheral blood (Table 1 and Fig 6B through D, F, H, and I). Peripheral blood smears of F8-injected SCID mice showed marked LGL lymphocytosis and neutrophilia (Fig 6H and I). Complete and differential blood counts of F8-injected SCID mice showed a significantly higher total white blood cell (WBC) count (11.1 ± 1.6 × 1012 WBCs/L) compared with normal mock-injected SCID mice (2.7 ± 1.3 × 1012 WBCs/L; Table 2). The leukocytosis in F8-injected SCID mice was primarily due to substantial increases in the LGL and neutrophil compartments. The F8-injected SCID mice also developed severe anemia, a very common hematologic feature observed in human NK-LGL leukemia.42 These engrafted mice also exhibited reticulocytosis and decreased hemoglobin levels (Table 2). To ensure that the expanded LGL cells observed in the F8-injected SCID mice were in fact the injected LGL cell line and not an expansion of T/NK cells from the SCID mice, a cell line from a peritoneal tumor was established (SC). This cell line was found to be morphologically and phenotypically identical to the original F8 LGL cell line. This similarity was confirmed by flow cytometric analysis, which showed that the F8 and SC cell lines possessed identical surface markers (CD3ε, TCR α/β, FcγR II/III, and IL-2Rβ; Fig 1). None of the six mock-injected SCID mice developed tumors during the same period (data not shown).

In contrast to the F8 LGL cell line, only one of six SCID mice injected with the IL-2–dependent LGL cell lines (f197 and m388) developed mild leukemia/lymphoma. The reason for this difference in SCID engraftment between the different LGL cell lines is unclear; however, it is possible that the SCID mice were unable to support the highly IL-2–dependent proliferation of the f197 and m388 cell lines.

Expression of adhesion molecules on Tax-transgenic LGL cell lines.Because nearly 100% of the mice from one of the GzmB-Tax transgenic founder lines (M9) develop peripheral tumors located on the tails, ears, and limbs (data not shown), flow cytometric analysis was performed on the established LGL cell lines from this founder line (f197 and m388) for the expression of cell surface adhesion molecules. As shown in Fig 7, both the f197 and m388 LGL cell lines expressed nearly identical levels of the same adhesion molecules, namely low levels of VLA-4, intermediate levels of ICAM-1, and high levels of LFA-1. Of note is the difference in adhesion molecule expression on the F8 (and SC) LGL cell line that was not derived from peripheral tail tumors, such as the f197 and m388 LGL cell lines, but was instead derived from a cervical lymph node. In particular, the F8 LGL cell line has virtually no LFA-1 expression and instead has an increase in VLA-4 and ICAM-1 expression. This differential expression of adhesion molecules may account for the difference in the anatomic origin of these LGL cell lines, as well as an explanation for the nearly 100% penetrance of peripheral tumor development in the M9 founder line. These observations are consistent with other data that have shown that both ICAM-1 and LFA-1 expression are increased in HTLV-1 infected cells,43-45 most likely due to an increase in transactivation by Tax.46 

Fig. 7.

Flow cytometric analysis of adhesion molecules on LGL cell lines. Reactivity of antibodies against adhesion molecules is indicated with solid lines. Reactivity of a control antibody, indicated by dashed lines, is depicted in all panels.

Fig. 7.

Flow cytometric analysis of adhesion molecules on LGL cell lines. Reactivity of antibodies against adhesion molecules is indicated with solid lines. Reactivity of a control antibody, indicated by dashed lines, is depicted in all panels.

Close modal

In this study, we have shown that targeted expression of Tax to the T/NK-cell compartment using the human GzmB promoter results in the development of immortalized LGL cells of T/NK-cell lineages. Importantly, each individual transgenic mouse develops morphologically similar peripheral tumors; however, each individual tumor is comprised of phenotypically distinct immortalized cell types (eg, CD3CD4CD8, CD3+CD4CD8, or CD3+CD4CD8+). These results suggest that, although Tax expression is sufficient for the predisposition of immortalization, various additional hits/mutations are required for tumor formation and leukemia. Other cellular proteins that have been suggested to play an important role in HTLV immortalization, such as inactivation of the p53 tumor-suppressor protein47,48 and activation of the Jak/Stat signal transduction pathway,49 50 are currently under investigation.

Fig. 8.

Hypothetical model for the role of cytokines in the process of peripheral tumor development in GzmB-Tax transgenic mice. This model would suggest that there is a need for an initial injury to peripheral skin tissue (eg, ear puncture, tail clipping, and fighting). This injury would subsequently start an inflammatory response by skin epithelial cells (eg, keratinocytes [KC]) that release inflammatory cytokines such as IL-1α and IL-1β. These inflammatory cytokines could then act on ECs of nearby vessels to cause the extravasation of circulating LGLs and neutrophils (PMN) by upregulating adhesion molecules such as ICAM-1 and VCAM-1 on ECs. Circulating LGLs also constitutively express adhesion molecules on their surface (LFA-1 and VLA-4), which promote their binding to activated ECs and extravasation. Once at the site of inflammation, these LGLs would become activated and release cytokines such as GM-CSF, which could then cause a systemic increase in PMN production and PMN extravasation into the tumors. In addition, these LGLs produce large quantities of IFNγ that could potentially initiate/exacerbate this inflammatory process by upregulating the expression of adhesion molecules such as ICAM-1 and VCAM-1 at this peripheral site.

Fig. 8.

Hypothetical model for the role of cytokines in the process of peripheral tumor development in GzmB-Tax transgenic mice. This model would suggest that there is a need for an initial injury to peripheral skin tissue (eg, ear puncture, tail clipping, and fighting). This injury would subsequently start an inflammatory response by skin epithelial cells (eg, keratinocytes [KC]) that release inflammatory cytokines such as IL-1α and IL-1β. These inflammatory cytokines could then act on ECs of nearby vessels to cause the extravasation of circulating LGLs and neutrophils (PMN) by upregulating adhesion molecules such as ICAM-1 and VCAM-1 on ECs. Circulating LGLs also constitutively express adhesion molecules on their surface (LFA-1 and VLA-4), which promote their binding to activated ECs and extravasation. Once at the site of inflammation, these LGLs would become activated and release cytokines such as GM-CSF, which could then cause a systemic increase in PMN production and PMN extravasation into the tumors. In addition, these LGLs produce large quantities of IFNγ that could potentially initiate/exacerbate this inflammatory process by upregulating the expression of adhesion molecules such as ICAM-1 and VCAM-1 at this peripheral site.

Close modal

We have also shown successful engraftment of a Tax-immortalized, IL-2–independent LGL cell line into SCID mice, resulting in the development of leukemia and lymphoma. The observed leukemia and lymphoma in these engrafted SCID mice closely resembled the pathological and phenotypical characteristics of the lymphoproliferative disorder that occurred in the original GzmB-Tax transgenic mice.25 The similarities between these two murine models included respiratory complications due to infiltration of LGLs, hepatomegaly due to LGL infiltration of the sinusoids, LGL infiltration of the lymph nodes and spleen, expansion of peripheral LGLs and neutrophils, reticulocytosis, and anemia. Of intrigue was the finding that the IL-2–dependent LGL cell lines were essentially incapable of being transferred to SCID mice, although the reason for this observation is unclear. Several possible explanations include (1) the inadequacy of SCID mice to support cytokine (IL-2) -dependent cell lines, (2) the lack of additional mutations in these IL-2–dependent cell lines needed for complete transformation, or (3) that these IL-2–dependent cell lines express certain surface antigens that are recognized by SCID NK cells that mediate their destruction. Recently, Stewart et al51 have reported the observation that HTLV-I–infected cell lines that lack viral gene expression are able to form tumors when inoculated into SCID mice, whereas HTLV-I–infected cells that do express viral gene products fail to form tumors. Such findings would support the difference in tumorigenicity between the IL-2–independent F8 cell line, which has lost detectable levels of Tax expression, and the IL-2–dependent f197 and m388 cell lines, which still express high levels of Tax (Fig 5). The first and third explanations are currently being investigated by coinjecting into SCID mice either recombinant human IL-2 or an NK-cell neutralizing antibody (anti–asialo-GM1) with the IL-2–dependent cell lines.

Although a definitive link between LGL leukemia and HTLV infection has yet to be established,6,52-55 the clinical features of human LGL leukemia are quite similar to those observed in the two murine models described in this report. These features include LGL leukocytosis, severe anemia, hepatosplenomegaly, bone marrow LGL infiltration, and lymphadenopathy. However, these murine models develop peripheral neutrophilia, a hematologic feature that is not common in human LGL patients. Instead, these patients tend to develop mild neutropenia.42 One plausible explanation for this difference may be due to the high level of GM-CSF expression in the GzmB-Tax transgenic mice, which is consistent with previous observations of peripheral neutrophilia and splenomegaly in mice injected with recombinant murine GM-CSF.56 Similar peripheral neutrophilia and splenomegaly were also reported in an earlier Tax transgenic mouse model that developed neurofibromas, which was shown to correspond to GM-CSF production by the transformed perineural fibroblasts.57 GM-CSF production in this model, as well as in our GzmB-Tax transgenic model, is most likely due to the ability of Tax to specifically transactivate the GM-CSF promoter.14,33 Of note, neutrophilia is a common clinical finding in ATL patients, which may be related to GM-CSF production by ATL cells.58 

In addition, we have also shown that there is a similar expression profile of certain cytokines (eg, IL-1α, IL-1β, GM-CSF, and IFNγ) among different GzmB-Tax transgenic mice. This conserved cytokine profile from two different transgenic mice suggests that these cytokines may play a critical role in the observed pathogenesis. Given the initial high dependence of the derived LGL cell lines for IL-2, it was surprising that we did not observe production of IL-2 in the peripheral tumors. This observation was further explored by crossing the Tax-transgenic mice with IL-2 knockout (KO) mice.59 The GzmB-Tax/IL-2 KO mice showed no differences in tumor development, progression, or phenotype from the original GzmB-Tax model (data not shown). These results are consistent with our failure to detect IL-2 message in vivo, but they do not rule out the possibility of paracrine IL-2 production or the role of IL-15.

As suggested in Fig 8, a model for the development of these peripheral tumors would include trauma to peripheral regions such as the tail, ears, and limbs, due to activities like tail clipping, ear puncture, and fighting, which leads to an inflammatory response. These peripheral sites are the most common areas (>90%) for the formation of primary tumors (data not shown). Inflammatory cytokines from epithelial cells, such as IL-1α and IL-1β, could potentially act on nearby endothelial cells (ECs) to induce expression of adhesion molecules such as ICAM-1 and VCAM-1. Circulating LGLs seem to constitutively express adhesion molecules on their surface (LFA-1 and VLA-4), which could promote their binding to activated ECs and eventual extravasation. Once at this site of inflammation, these LGLs release GM-CSF, which is most likely the cause of the observed peripheral neutrophilia and polymorphonuclear leukocyte (PMN) recruitment into the tumors. High levels of IFNγ production by these LGL cells may potentially initiate and/or exacerbate this inflammatory process by upregulating the expression of adhesion molecules (ICAM-1 and VCAM-1) on ECs as well as epithelial cells such as keratinocytes.60 In fact, preliminary experiments suggest that injecting a neutralizing antibody against IFNγ into these transgenic mice significantly reduces the progression of these peripheral tumors. Another explanation currently being explored for the importance of IFNγ in this transgenic model is whether it is playing a protective role in preventing cellular apoptosis.61 

Indeed, recent transgenic models that use either the human GzmB or GzmH promoters driving expression of the simian virus 40 large T antigen (SVlgT) also develop very similar T/NK-cell lymphomas.62 However, these transgenic mice do not develop the peripheral tumors or neutrophilia observed in the GzmB-Tax transgenic mice. These data suggest that Tax expression may be responsible for these observed phenotypes, possibly through dysregulated cytokine and/or adhesion molecule production. Data to support this hypothesis include the inability to detect IFNγ or GM-CSF production in the majority of LGL cell lines derived from GzmH-SVlgT transgenic mice (data not shown; MacIvor, Grossman, Ratner, and Ley, personal communication, January, 1997).

Overall, both the GzmB-Tax transgenic and LGL cell line-engrafted SCID models provide useful tools to examine the complex cellular mechanisms that are involved in the immortalization process by HTLV infection. In particular, these models have been shown to be useful in the dissection of the role of cytokine expression in the observed pathogenesis. Such examinations may prove to be useful in understanding the disease progression of HTLV-1–associated diseases such as ATL, TSP, and cutaneous T-cell lymphoma (mycosis fungoides), in which cytokine and adhesion molecule expression have been speculated to play an important role in organ infiltration and disease development.33,36,43,45,60 62-65 

Transgenic mouse development was supported by a National Institute of Child Health and Human Development contract to DNX, Inc. The authors thank Dr Marie LaRegina, Niecey Hinkle, and Paula Klender at the Division of Comparative Medicine for their help in peripheral blood analysis, and Dr Timothy J. Ley for critical review of this manuscript.

Supported by Grants No. CA63417 and 5T32 HL07088.

Address reprint requests to Lee Ratner, MD, PhD, Washington University School of Medicine, Division of Molecular Oncology, Box 8069, 660 S Euclid Ave, St Louis, MO 63110.

1
Gallo
RC
Kalyanaraman
VS
Sarngadharan
MG
Sliski
A
Vonderheid
EC
Maeda
M
Nakao
Y
Yamada
K
Ito
Y
Gutensohn
N
Murphy
S
Bunn
PA Jr
Catovsky
D
Greaves
MF
Blayney
DW
Blattner
W
Jarrett
WFH
zur Hausen H
Seligmann
M
Brouet
JC
Haynes
BF
Jegasothy
BV
Jaffe
E
Cossman
J
Broder
S
Fisher
RI
Golde
DW
Robert-Guroff
M
Association of the human type C retrovirus with a subset of adult T-cell Cancers.
Cancer Res
43
1983
3892
2
Gessain
A
Barin
F
Vernant
JC
Gout
O
Maurs
L
Calender
A
de The
G
Antibodies to human T-lymphotropic virus type-I in patients with tropical spastic paraparesis.
Lancet
2
1985
407
3
Nishioka
K
Maruyama
I
Sato
K
Kitajima
I
Nakajima
Y
Osame
M
Chronic inflammatory arthropathy associated with HTLV-I (letter).
Lancet
1
1989
441
4
Mochizuki
M
Watanabe
T
Yamaguchi
K
Takatsuki
K
Yoshimura
K
Shirao
M
Nakashima
S
Mori
S
Araki
S
Miyata
N
HTLV-I uveitis: A distinct clinical entity caused by HTLV-I.
Jpn J Cancer Res
83
1992
236
5
Pancake
BA
Zucker-Franklin
D
Coutavas
EE
The cutaneous T cell lymphoma, mycosis fungoides, is a human T cell lymphotropic virus-associated disease. A study of 50 patients.
J Clin Invest
95
1995
547
6
Loughran
TP Jr
Coyle
T
Sherman
MP
Starkebaum
G
Ehrlich
GD
Ruscetti
FW
Poiesz
BJ
Detection of human T-cell leukemia/lymphoma virus, type II, in a patient with large granular lymphocyte leukemia.
Blood
80
1992
1116
7
Martin
MP
Biggar
RJ
Hamlin-Green
G
Staal
S
Mann
D
Large granular lymphocytosis in a patient infected with HTLV-II.
AIDS Res Hum Retroviruses
9
1993
715
8
Franchini
G
Molecular mechanisms of human T-cell leukemia/lymphotropic virus type I infection.
Blood
86
1995
3619
9
Cann
AJ
Rosenblatt
JD
Wachsman
W
Shah
NP
Chen
ISY
Identification of the gene responsible for human T-cell leukaemia virus transcriptional regulation.
Nature
318
1985
571
10
Duyao
MP
Kessler
DJ
Spicer
DB
Bartholomew
C
Cleveland
JL
Siekevitz
M
Sonenshein
GE
Transactivation of the c-myc promoter by human T cell leukemia virus type 1 tax is mediated by NF kappa B.
J Biol Chem
267
1992
16288
11
Ratner
L
Thielan
B
Collins
T
Sequences of the 5′ portion of the human c-sis gene: Characterization of the transcriptional promoter and regulation of expression of the protein product by 5′ untranslated mRNA sequences.
Nucleic Acids Res
15
1987
6017
12
Fujii
M
Niki
T
Mori
T
Matsuda
T
Matsui
M
Nomura
N
Seiki
M
HTLV-1 Tax induces expression of various immediate early serum responsive genes.
Oncogene
6
1991
1023
13
Siekevitz
M
Feinberg
MB
Holbrook
N
Wong-Staal
F
Greene
WC
Activation of interleukin 2 and interleukin 2 receptor (Tac) promoter expression by the trans-activator (tat ) gene product of human T-cell leukemia virus, type I.
Proc Natl Acad Sci USA
84
1987
5389
14
Himes
SR
Coles
LS
Katsikeros
R
Lang
RK
Shannon
MF
HTLV-1 tax activation of the GM-CSF and G-CSF promoters requires the interaction of NF-kB with other transcription factor families.
Oncogene
8
1993
3189
15
Tanaka
A
Takahashi
C
Yamaoka
S
Nosaka
T
Maki
M
Hatanaka
M
Oncogenic transformation by the tax gene of human T-cell leukemia virus type I in vitro.
Proc Natl Acad Sci USA
87
1990
1071
16
Inoue
H
Yamaoka
S
Imamura
M
Hatanaka
M
Suppression of the transformed phenotype in hybrids of human T-cell leukemia virus type I tax-transformed rat fibroblasts and normal human fibroblasts.
Exp Cell Res
215
1994
68
17
Pozzatti
R
Vogel
J
Jay
G
The human T-lymphotropic virus type I tax gene can cooperate with the ras oncogene to induce neoplastic transformation of cells.
Mol Cell Biol
10
1990
413
18
Grassmann
R
Berchtold
S
Radant
I
Alt
M
Fleckenstein
B
Sodroski
JG
Haseltine
WA
Ramstedt
U
Role of human T-cell leukemia virus type 1 X region proteins in immortalization of primary human lymphocytes in culture.
J Virol
66
1992
4570
19
Ratner L: Animal models of HTLV-I Infection, in Höllsberg P, Hafler DA (eds): Human T-Cell Lymphotropic Virus Type I, vol 1. New York, NY, Wiley, 1996, p 287
20
Nerenberg
M
Hinrichs
SH
Reynolds
RK
Khoury
G
Jay
G
The tat gene of human T-lymphotropic virus type 1 induces mesenchymal tumors in transgenic mice.
Science
237
1987
1324
21
Iwakura
Y
Tosu
M
Yoshida
E
Takiguchi
M
Sato
K
Kitajima
I
Nishioka
K
Yamamoto
K
Takeda
T
Hatanaka
M
Yamamoto
H
Sekiguchi
T
Induction of inflammatory arthropathy resembling rheumatoid arthritis in mice transgenic for HTLV-I.
Science
253
1991
1026
22
Furuta
Y
Aizawa
S
Suda
Y
Ikawa
Y
Kishimoto
H
Asano
Y
Tada
T
Hikikoshi
A
Yoshida
M
Seiki
M
Thymic atrophy characteristic in transgenic mice that harbor pX genes of human T-cell leukemia virus type I.
J Virol
63
1989
3185
23
Nerenberg
MI
Minor
T
Price
J
Ernst
DN
Shinohara
T
Schwarz
H
Transgenic thymocytes are refractory to transformation by the human T-cell leukemia virus type I tax gene.
J Virol
65
1991
3349
24
Benvenisty
N
Ornitz
DM
Bennett
GL
Sahagan
BG
Kuo
A
Cardiff
RD
Leder
P
Brain tumours and lymphomas in transgenic mice that carry HTLV-I LTR/c-myc and Ig/tax genes.
Oncogene
7
1992
2399
25
Grossman
WJ
Kimata
JT
Wong
FH
Zutter
M
Ley
TJ
Ratner
L
Development of leukemia in mice transgenic for the tax gene of human T-cell leukemia virus type I.
Proc Natl Acad Sci USA
92
1995
1057
26
Montgomery
RA
Dallman
MJ
Analysis of cytokine gene expression during fetal thymic ontogeny using the polymerase chain reaction.
J Immunol
147
1991
554
27
Langton
BC
Sliwkowski
M
Tran
KV
Knapp
S
Keitelman
E
Smith
C
Wallingford
S
Liu
H-L
Ralston
JS
Brandis
J
Coats
S
Development and characterization of monoclonal antibodies to the HTLV-I Tax (p 40X) protein.
Med Virol
8
1988
295
28
Hansen
MB
Nielson
SE
Berg
K
Re-examination and further development of a precise and rapid dye method for measuring cell growth/cell kill.
J Immunol Methods
119
1989
203
29
Maruyama
M
Shibuya
H
Harada
H
Hatakeyama
M
Seiki
M
Fujita
T
Inoue
J
Yoshida
M
Taniguchi
T
Evidence for aberrant activation of the interleukin-2 autocrine loop by HTLV-1-encoded p40x and T3/Ti complex triggering.
Cell
48
1987
343
30
Knowles
DM
Immunophenotypic and antigen receptor gene rearrangement analysis in T cell neoplasia.
Am J Pathol
134
1989
761
31
Richardson
JH
Edwards
AJ
Cruickshank
JK
Rudge
P
Dalgleish
AG
In vivo cellular tropism of human T-cell leukemia virus type 1.
J Virol
64
1990
5682
32
Hanson
RD
Sclar
GM
Kanagawa
O
Ley
TJ
The 5′-flanking region of the human CGL-1/granzyme B gene targets expression of a reporter gene to activated T-lymphocytes in transgenic mice.
J Biol Chem
266
1991
24433
33
Sawada
M
Suzumura
A
Kondo
N
Marunouchi
T
Induction of cytokines in glial cells by trans activator of human T-cell lymphotropic virus type I.
FEBS Lett
313
1992
47
34
Wano
Y
Hattori
T
Matsuoka
M
Takatsuki
K
Chua
AO
Gubler
U
Greene
WC
Interleukin 1 gene expression in adult T-cell leukemia.
J Clin Invest
80
1987
911
35
Yamashita
I
Katamine
S
Moriuchi
R
Nakamura
Y
Miyamoto
T
Eguchi
K
Nagataki
S
Transactivation of the human interleukin-6 gene by human T-lymphotropic virus type 1 Tax protein.
Blood
84
1994
1573
36
Sagawa
K
Mochizuki
M
Masuoka
K
Katagiri
K
Katayama
T
Maeda
T
Tanimoto
A
Sugita
S
Watanabe
T
Itoh
K
Immunopathological mechanisms of human T cell lymphotropic virus type 1 (HTLV-I) uveitis. Detection of HTLV-I-infected T cells in the eye and their constitutive cytokine production.
J Clin Invest
95
1995
852
37
Kim
SJ
Kehrl
JH
Burton
J
Tendler
CL
Jeang
KT
Danielpour
D
Thevenin
C
Kim
KY
Sporn
MB
Roberts
AB
Transactivation of the transforming growth factor beta 1 (TGF-beta 1) gene by human T lymphotropic virus type 1 tax: A potential mechanism for the increased production of TGF-beta 1 in adult T cell leukemia.
J Exp Med
172
1990
121
38
Kurosawa
S
Matsuzaki
G
Harada
M
Ando
T
Nomoto
K
Early appearance and activation of natural killer cells in tumor-infiltrating lymphoid cells during tumor development.
Eur J Immunol
23
1993
1029
39
Cuturi
MC
Anegon
I
Sherman
F
Loudon
R
Clark
SC
Perussia
B
Trinchieri
G
Production of hematopoietic colony-stimulating factors by human natural killer cells.
J Exp Med
169
1989
569
40
Murphy
WJ
Keller
JR
Harrison
CL
Young
HA
Longo
DL
Interleukin-2–activated natural killer cells can support hematopoiesis in vitro and promote marrow engraftment in vivo.
Blood
80
1992
670
41
Brown
DA
Nelson
FB
Reinherz
EL
Diamond
DJ
The human interferon-gamma gene contains an inducible promoter that can be transactivated by tax I and II.
Eur J Immunol
21
1991
1879
42
Loughran
TP Jr
Clonal diseases of large granular lymphocytes [see comments].
Blood
82
1993
1
43
Fukudome
K
Furuse
M
Fukuhara
N
Orita
S
Imai
T
Takagi
S
Nagira
M
Hinuma
Y
Yoshie
O
Strong induction of ICAM-1 in human T cells transformed by human T-cell-leukemia virus type 1 and depression of ICAM-1 or LFA-1 in adult T-cell-leukemia-derived cell lines.
Int J Cancer
52
1992
418
44
Imai
T
Tanaka
Y
Fukudome
K
Takagi
S
Araki
K
Yoshie
O
Enhanced expression of LFA-3 on human T-cell lines and leukemic cells carrying human T-cell-leukemia virus type 1.
Int J Cancer
55
1993
811
45
Uchiyama
T
Ishikawa
T
Imura
A
Adhesion properties of adult T cell leukemia cells.
Leuk Lymphoma
16
1995
407
46
Tanaka
Y
Fukudome
K
Hayashi
M
Takagi
S
Yoshie
O
Induction of ICAM-1 and LFA-3 by Tax1 of human T-cell leukemia virus type 1 and mechanism of down-regulation of ICAM-1 or LFA-1 in adult-T-cell-leukemia cell lines.
Int J Cancer
60
1995
554
47
Yamato
K
Oka
T
Hiroi
M
Iwahara
Y
Sugito
S
Tsuchida
N
Miyoshi
I
Aberrant expression of the p53 tumor suppressor gene in adult T-cell leukemia and HTLV-I-infected cells.
Jpn J Cancer Res
84
1993
4
48
Cesarman
E
Chadburn
A
Inghirami
G
Gaidano
G
Knowles
DM
Structural and functional analysis of oncogenes and tumor suppressor genes in adult T-cell leukemia/lymphoma shows frequent p53 mutations.
Blood
80
1992
3205
49
Migone
TS
Lin
JX
Cereseto
A
Mulloy
JC
O'Shea
JJ
Franchini
G
Leonard
WJ
Constitutively activated Jak-STAT pathway in T cells transformed with HTLV-I.
Science
269
1995
79
50
Xu
X
Kang
SH
Heidenreich
O
Okerholm
M
O'Shea
JJ
Nerenberg
MI
Constitutive activation of different Jak tyrosine kinases in human T cell leukemia virus type 1 (HTLV-1) tax protein or virus-transformed cells.
J Clin Invest
96
1995
1548
51
Stewart
SA
Feuer
G
Jewett
A
Lee
FV
Bonavida
B
Chen
ISY
HTLV-1 gene expression in adult T-cell leukemia cells elicits an NK cell response in vitro and correlates with cell rejection in SCID mice.
Virology
226
1996
167
52
Levitt
LJ
Reyes
GR
Moonka
DK
Bensch
K
Miller
RA
Engleman
EG
Human T cell leukemia virus-I-associated T-suppressor cell inhibition of erythropoiesis in a patient with pure red cell aplasia and chronic T gamma-lymphoproliferative disease.
J Clin Invest
81
1988
538
53
Loughran
TP Jr
Kidd
P
Poiesz
BJ
Familial occurrence of LGL leukaemia.
Br J Haematol
87
1994
199
54
Loughran
TP Jr
Sherman
MP
Ruscetti
FW
Frey
S
Coyle
T
Montagna
RA
Jones
B
Starkebaum
G
Poiesz
BJ
Prototypical HTLV-I/II infection is rare in LGL leukemia.
Leuk Res
18
1994
423
55
Heneine
W
Chan
WC
Lust
JA
Sinha
SD
Zaki
SR
Khabbaz
RF
Kaplan
JE
HTLV-II infection is rare in patients with large granular lymphocyte leukemia [letter].
J AIDS
7
1994
736
56
Metcalf
D
Begley
CG
Williamson
DJ
Nice
EC
Lamarter
JD
Mermod
J-J
Thatcher
D
Schmidt
A
Hemopoietic responses in mice injected with purified recombinant murine GM-CSF.
Exp Hematol
15
1987
1
57
Green
JE
Begley
CG
Wagner
DK
Waldmann
TA
Jay
G
Trans activation of granulocyte-macrophage colony-stimulating factor and the interleukin-2 receptor in transgenic mice carrying the human T-lymphotropic virus type 1 tax gene.
Mol Cell Biol
9
1989
4731
58
Yamamoto
S
Hattori
T
Asou
N
Nishimura
H
Kawano
F
Yodoi
J
Takatsuki
K
Absolute neutrophilia in adult T cell leukemia.
Jpn J Cancer Res
77
1986
858
59
Kundig
TM
Schorle
H
Bachmann
MF
Hengartner
H
Zinkernagel
RM
Horak
I
Immune responses in interleukin-2-deficient mice.
Science
262
1993
1059
60
Jin
F
Iwatsuki
K
Takigawa
M
Yamada
M
Induction of intercellular adhesion molecule-1 and adherence of HTLV-1-infected T-cells to cultured keratinocytes.
J Dermatol
21
1994
172
61
Clerici
M
Sarin
A
Coffman
RL
Wynn
TA
Blatt
SP
Hendrix
CW
Wolf
SF
Shearer
GM
Henkart
PA
Type 1/type 2 cytokine modulation of T-cell programmed cell death as a model for human immunodeficiency virus pathogenesis.
Proc Natl Acad Sci USA
91
1994
11811
62
Umehara
F
Izumo
S
Ronquillo
AT
Matsumuro
K
Sato
E
Osame
M
Cytokine expression in the spinal cord lesions in HTLV-I-associated myelopathy.
J Neuropathol Exp Neurol
53
1994
72
63
Kodaka
T
Uchiyama
T
Umadome
H
Uchino
H
Expression of cytokine mRNA in leukemic cells from adult T cell leukemia patients.
Jpn J Cancer Res
80
1989
531
64
Ichinose
K
Nakamura
T
Nishiura
Y
Nagasato
K
Ohishi
K
Watanabe
H
Fujita
A
Kurouji
K
Tsujihata
M
Nagataki
S
Characterization of adherent T cells to human endothelial cells in patients with HTLV-I-associated myelopathy.
J Neurol Sci
122
1994
204
65
Gallo RC, Thomson MM: Introduction, in Höllsberg P, Hafler DA (eds): Human T-Cell Lymphotropic Virus Type I, vol 1. New York, NY, Wiley, 1996, p 1
Sign in via your Institution