Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cancer Gene Ther ; 6(5): 447-55, 1999.
Article in English | MEDLINE | ID: mdl-10505855

ABSTRACT

The transfer of genes encoding cytokines into tumor cells has emerged as a new strategy to increase in vivo host reactivity to a variety of tumors. Because gene transfer into tumor cells cannot be easily applied in the clinical setting, we have developed an experimental model of gene transfer into fibroblasts and examined the capacity of these engineered cells to elicit an antitumor immune response. Interleukin-12 (IL-12) is a heterodimeric cytokine with pleiotropic activities presenting strong antitumor and antimetastatic effects in murine models. A bicistronic retroviral vector was constructed that contained the cDNAs encoding both chains (p40 and p35) of murine IL-12 separated by an internal ribosomal entry site sequence. Syngeneic cutaneous fibroblasts obtained from newborn mice and transduced to secrete either IL-12 or IL-2 were injected subcutaneously with B16F0 or B16F1 melanoma cells. The time of tumor occurrence and overall survival of mice were significantly prolonged when B16F1 cells were coinjected with cytokine-producing fibroblasts compared with B16F1 alone or B16F1 together with unmanipulated fibroblasts. Systemic effects were seen in the mice injected with either IL-2- or IL-12-secreting fibroblasts, with the highest proliferation capability and interferon-gamma production observed in vitro from splenocytes from recipients of IL-2-secreting fibroblasts. Injection of IL-2-secreting fibroblasts or coinjection of IL-2- and IL-12-producing fibroblasts resulted in a significant increase of survival in the B16F0 model; in some cases, complete disease eradication was observed. These results suggest that cutaneous fibroblasts represent a target of choice for gene transfer and would be useful in the treatment of minimal residual disease in humans.


Subject(s)
Gene Transfer Techniques , Interleukin-12/genetics , Interleukin-2/genetics , Melanoma, Experimental/immunology , Retroviridae/genetics , Animals , Base Sequence , DNA Primers , Fibroblasts/metabolism , Genetic Therapy , Genetic Vectors , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL
2.
Blood ; 92(6): 2003-11, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9731058

ABSTRACT

flt3/flk-2 ligand (FL) is a cytokine that exhibits synergistic activities in combination with other early acting factors on subpopulations of hematopoietic stem/progenitor cells. In addition to normal hematopoietic precursors, expression of the FL receptor, flt3R, has been frequently demonstrated on the blast cells from patients with acute B-lineage lymphoblastic, myeloid, and biphenotypic (also known as hybrid or mixed) leukemias. Because many of these leukemic cell types express FL, the possibility has been raised that altered regulation of FL-mediated signaling might contribute to malignant transformation or expansion of the leukemic clone. In humans, FL is predominantly synthesized as a transmembrane protein that must undergo proteolytic cleavage to generate a soluble form. To investigate the consequences of constitutively expressing the analogous murine FL isoform in murine hematopoietic stem/progenitor cells, lethally irradiated syngeneic mice (18 total) were engrafted with post-5-fluorouracil-treated bone marrow cells transduced ex vivo with a recombinant retroviral vector (MSCV-FL) encoding murine transmembrane FL. Compared with control mice (8 total), MSCV-FL mice presented with a mild macrocytic anemia but were otherwise healthy for more than 5 months posttransplant (until 22 weeks). Subsequently, all primary MSCV-FL recipients observed for up to 1 year plus 83% (20 of 24) of secondary MSCV-FL animals that had received bone marrow from asymptomatic primary hosts reconstituted for 4 to 5 months developed transplantable hematologic malignancies (with mean latency periods of 30 and 23 weeks, respectively). Phenotypic and molecular analyses indicated that the tumor cells expressed flt3R and displayed B-cell and/or myeloid markers. These data, establishing that dysregulated expression of FL in primitive hematopoietic cells predisposes flt3R+ precursors to leukemic transformation, underscore a potential role of this cytokine/receptor combination in certain human leukemias.


Subject(s)
Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Leukemia, Experimental/genetics , Membrane Proteins/biosynthesis , Animals , Bone Marrow/metabolism , Clone Cells , Disease Susceptibility , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors/genetics , Leukemia, Experimental/etiology , Leukemia, Experimental/pathology , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Recombinant Proteins/biosynthesis , Tumor Cells, Cultured
3.
Blood ; 92(3): 877-87, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9680355

ABSTRACT

Primitive and definitive erythropoiesis represent distinct hematopoietic programs that differ with respect to stage of development, transcriptional control, and growth regulation. Although these differences have been recognized for some time, the relationship of the two erythroid lineages to each other is not well established. We have used a model system based on the hematopoietic development of embryonic stem (ES) cells in culture to investigate the origins of the earliest hematopoietic populations. Using ES cells transduced with a retrovirus that overexpresses the HOX11 gene, we have established factor-dependent hematopoietic cell lines that represent novel stages of embryonic hematopoiesis. Analysis of three of these cell lines indicates that they differ with respect to cytokine responsiveness, cell surface markers, and developmental potential. Two of the cell lines, EBHX1 and EBHX11, display the unique capacity to generate both primitive and definitive erythroid progeny as defined by morphology and expression of betaH1 and betamajor globin. The third line, EBHX14, has definitive erythroid and myeloid potential, but is unable to generate cells of the primitive erythroid lineage. Analysis of the cytokine responsiveness of the two lines with primitive erythroid potential has indicated that exposure to leukemia inhibitory factor (LIF) results in the upregulation of betaH1 and a change in cellular morphology to that of primitive erythrocytes. These findings are the first demonstration of a clonal cell line with primitive and definitive hematopoietic potential and support the interpretation that these lineages may arise from a common precursor in embryonic life. In addition, they suggest that LIF could play a role in the regulation of primitive erythropoiesis.


Subject(s)
Erythropoiesis , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/physiology , Interleukin-6 , Oncogene Proteins/physiology , Animals , CHO Cells , Carcinoma, Embryonal/pathology , Cell Differentiation , Cell Lineage , Cells, Cultured , Cricetinae , Cricetulus , Culture Media, Conditioned/pharmacology , Endothelial Growth Factors/pharmacology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Flow Cytometry , Gene Expression Regulation, Developmental/drug effects , Globins/biosynthesis , Globins/genetics , Growth Inhibitors/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/classification , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Leukemia Inhibitory Factor , Lymphokines/pharmacology , Multiple Myeloma/pathology , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Recombinant Fusion Proteins/physiology , Stem Cell Factor/pharmacology , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
5.
Cancer Gene Ther ; 4(3): 167-75, 1997.
Article in English | MEDLINE | ID: mdl-9171935

ABSTRACT

The combination of immunotherapy with conventional treatments such as radio- and chemotherapy may be necessary to eradicate minimal residual disease. Interleukin 12 (IL-12) is a heterodimeric cytokine composed of two subunits, p40 and p35. Coordinate expression of the IL-12 p40 and p35 genes in several solid tumor models has been found to induce strong and specific antitumor immune responses. In the interest of obtaining high level IL-12 expression in leukemia/lymphoma cells for use as vaccines in cancer immunotherapy, we evaluated three IL-12 retroviral vector designs based on the murine stem cell virus (MSCV) vector which efficiently transduces functional genes into normal hematopoietic cells. MSCVpac-mlL-12 and MIPV-mIL-12 contain an encephalomyocarditis virus internal ribosome entry site for internal translation of bicistronic mRNA transcripts, while MDCVpac-mIL-12 carries an expression cassette in the U3 region of the 3' long terminal repeat. We found that the MSCVpac-mIL-12 vector directed robust expression of both p40 and p35 genes in several murine tumor cell lines of hematopoietic origin, including a T-cell lymphoma, a B-cell lymphoma, and a plasmacytoma/myeloma. In contrast, genomic instability or promoter interference hampered p40 gene expression in cells transduced with the MIPV-mIL-12 and MDCVpac-mIL-12 vectors, respectively. These findings provide the basis for the design of IL-12 retroviral vectors for the treatment of hematologic malignancies in humans.


Subject(s)
Genetic Therapy , Genetic Vectors , Interleukin-12/genetics , Retroviridae/genetics , Acetyltransferases/genetics , Animals , Evaluation Studies as Topic , Gene Transfer Techniques , Genes , Hematopoietic Stem Cells , Interleukin-12/immunology , Interleukin-12/therapeutic use , Interleukin-9 , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/therapy , Lymphoma, T-Cell/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Tumor Cells, Cultured
6.
Cancer Res ; 57(2): 337-45, 1997 Jan 15.
Article in English | MEDLINE | ID: mdl-9000579

ABSTRACT

The HOX11/TCL3 homeobox gene was identified at the breakpoint region in pediatric T-cell acute lymphoblastic leukemia harboring 10q24 chromosomal translocations. We previously reported that primary murine bone marrow cells transduced ex vivo with a recombinant HOX11-containing retrovirus, MSCV-HOX11, gave rise to cell lines at high frequency having characteristics of early myeloid cells. Cell lines were also established from the bone marrow and spleen of transplant recipients sacrificed 5 months after engraftment with MSCV-HOX11-transduced bone marrow cells. These latter lines, which exhibited a more differentiated myelomonocytic phenotype, harbored proviruses encoding a smaller HOX11 protein. None of the mice that received HOX11-expressing bone marrow cells or myeloid cell lines developed leukemia during 6-month observation periods. Here, we report that two bone marrow transplant recipients eventually developed T-cell acute lymphoblastic leukemia-like malignancies at 7 and 12 months posttransplant, indicating that progression to a fully malignant state required additional mutations. One tumor synthesized full-length HOX11 whereas the other expressed the smaller version of the protein. The smaller HOX11 protein suffered a carboxyl-terminal truncation. We subsequently constructed MSCV-based retroviral vectors expressing deleted forms of HOX11 and identified an amino-terminal region that was dispensible for generation of myeloid cell lines having a similar phenotype as those induced by full-length HOX11. We thus conclude that regions near the amino and carboxyl termini of HOX11 are not essential for transforming function, nor do they appear to determine the lineage or stage of differentiation of the target cell for transformation.


Subject(s)
Cell Transformation, Viral/genetics , Genes, Homeobox/physiology , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Leukemia, T-Cell/genetics , Oncogene Proteins/genetics , Retroviridae/genetics , Sequence Deletion , Animals , Bone Marrow Transplantation , Female , Leukemia, T-Cell/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Transfection , Tumor Cells, Cultured
7.
Gene Ther ; 4(10): 1061-8, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9415312

ABSTRACT

The efficient genetic modification of solid tumors in situ to stimulate therapeutic immune responses against them is currently under active investigation, but is not yet possible using existing gene transfer technologies. Thus, ex vivo/in vivo vaccination strategies have been proposed in which the patient's tumor is surgically excised, single cell suspensions are prepared, the therapeutic genes are introduced and then the gene-modified cells, after being gamma-irradiated, are injected back into the patient. However, even with high-efficiency gene delivery systems, this is a labor-intensive process. Moreover, it is often difficult to obtain sufficient numbers of gene-modified primary tumor cells during short-term culturing. On the other hand, extended in vitro passaging of primary tumor explants may alter their immunophenotypic properties. One approach to overcome these limitations would be to design universal vaccines consisting of standardized gene-transduced neoplastic cell lines or mixtures of gene-transduced cell lines to be combined with autologous tumor samples if available. Melanoma, which is notable for being one of the most immunogenic human malignancies, represents a cancer where shared tumor-associated antigens have been identified. We developed and analyzed several different retroviral vectors for their ability to stably express exogenous genes at high levels in a panel of melanoma cell lines. All vectors contained a reporter gene (nlslacZ) encoding beta-galactosidase with a nuclear localization signal and the neomycin phosphotransferase (neo) gene as selectable marker. One vector, DCCMV, which carried a bicistronic nlslacZ-neo transcriptional unit under the control of the human cytomegalovirus immediate-early promoter in the U3 region of its 3' LTR, was found to perform consistently better than the other vectors. The DCCMV vector, which is an extreme example of the double-copy class of retroviral vectors, was subsequently used to generate melanoma cell lines constitutively secreting human interleukin-6 or a soluble form of the human interleukin-6 receptor for potential use in a phase II clinical vaccine trial for the treatment of melanoma patients. The DCCMV vector design may also be useful in gene therapy applications where the intent is to implant polymer-encapsulated cell lines genetically engineered to stably express high levels of bioactive proteins.


Subject(s)
Cancer Vaccines/genetics , Gammaretrovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Melanoma/genetics , Blotting, Northern , Gene Expression , Genetic Engineering/methods , Humans , Interleukin-6/metabolism , Melanoma/immunology , Melanoma/therapy , Receptors, Interleukin-6/metabolism , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 93(19): 10297-302, 1996 Sep 17.
Article in English | MEDLINE | ID: mdl-8816794

ABSTRACT

Based on transplantation studies with bone marrow cultured under various conditions, a role of interleukin 11 (IL-11) in the self-renewal and/or the differentiation commitment of hematopoietic stem cells has been indicated. To better evaluate the in vivo effects of IL-11 on stem/progenitor cell biology, lethally irradiated mice were serially transplanted with bone marrow cells transduced with a defective retrovirus, termed MSCV-mIL-11, carrying the murine IL-11 (mIL-11) cDNA and the bacterial neomycin phosphotransferase (neo) gene. High serum levels (i.e., > 1 ng/ml) of mIL-11 in all (20/20) primary and 86% (12/14) of secondary long-term reconstituted mice, as well as 86% (12/14) of tertiary recipients examined at 6 weeks posttransplant, demonstrated persistence of vector expression subsequent to transduction of bone marrow precursors functionally definable as totipotent hematopoietic stem cells. In agreement with results obtained with human IL-11 in other myeloablation models, ectopic mIL-11 expression accelerated recovery of platelets, neutrophils, and, to some extent, total leukocytes while preferentially increasing peripheral platelet counts in fully reconstituted mice. When analyzed 5 months posttransplant, tertiary MSCV-mIL-11 recipients had a significantly greater percentage of G418-resistant colony-forming cells in their bone marrow compared with control MSCV animals. Collectively, these data show that persistent stimulation of platelet production by IL-11 is not detrimental to stem cell repopulating ability; rather, they suggest that IL-11 expression in vivo may have resulted in enhanced maintenance of the most primitive hematopoietic stem cell compartment. The prolonged expression achieved by the MSCV retroviral vector, despite the presence of a selectable marker, contrasts with the frequent transcriptional extinction observed with other retroviral vectors carrying two genes. These findings have potentially important implications for clinical bone marrow transplantation and gene therapy of the hematopoietic system.


Subject(s)
Bone Marrow Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Interleukin-11/physiology , Animals , Bone Marrow/radiation effects , Colony-Forming Units Assay , DNA, Complementary , Female , Gene Expression , Humans , Interleukin-11/biosynthesis , Interleukin-11/blood , Leukocyte Count , Mice , Mice, Inbred C57BL , Platelet Count , Recombinant Proteins/biosynthesis , Recombinant Proteins/blood , Retroviridae , Time Factors , Transfection , Transplantation, Isogeneic
10.
Oncogene ; 11(6): 1113-23, 1995 Sep 21.
Article in English | MEDLINE | ID: mdl-7566971

ABSTRACT

Although activating mutations in ras genes are the most common genetic abnormality in human hematologic malignancies, the role of ras mutations as an initiating event in leukemogenesis remains unclear. To assess the consequences of ectopic expression of an activated ras gene in normal hematopoietic cells in vivo, lethally irradiated mice were reconstituted with bone marrow cells infected with a mutant ras-containing retrovirus [murine stem cell virus (MSCV)-v-H-ras] based on the MSCV retroviral vector which efficiently transduces functional genes into hematopoietic stem/progenitor cells. Despite a marked myeloid leukocytosis detectable in the peripheral blood within 4 weeks of engraftment, none of 22 primary or secondary transplant recipients studied for longer periods of time presented with myeloid neoplasms. Instead, 18 of the MSCV-v-H-ras mice developed pre-T-cell thymic lymphomas and/or pre-B-cell lymphoblastic leukemia/lymphomas between 7 and 12 weeks post-transplantation. The pre-B and pre-T lymphoid tumors that arose in one animal were shown to harbor a common MSCV-v-H-ras provirus, indicating that the target cell for transformation was a bipotential lymphoid precursor. To more precisely examine the effects of activated ras expression on the behavior of hematopoietic progenitors, infected bone marrow cells were assayed in methylcellulose cultures under conditions favorable for growth of multilineage myeloid colonies or were passaged as bulk suspension cultures in the presence of various hematopoietic growth factors, including interleukin (IL)-3, IL-4, IL-6 and IL-7. MSCV-directed expression of v-H-ras selectively promoted the formation of large dense colonies comprised of monocyte-macrophages in methylcellulose cultures. When transferred to liquid cultures, the vast majority of the cells underwent terminal macrophage differentiation. By comparison, tumorigenic B-lymphoid and mixed lymphoid/myeloid cell lines were routinely established from the bulk suspension cultures, with cell lines of predominantly myeloid phenotype emerging only in IL-6-supplemented cultures. These results, considered together with previous findings, suggest that activating ras mutations could be an initiating genetic alteration in human acute lymphoblastic leukemia but are more likely to be a post-initiation change in human acute myeloid leukemia.


Subject(s)
Burkitt Lymphoma/etiology , Cell Transformation, Neoplastic , Genes, ras , Lymphoma/etiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Animals , Chimera , Female , Gene Rearrangement , Hematopoiesis , Hematopoietic Stem Cells/virology , Mice , Mice, Inbred BALB C , Mutation , Proviruses/genetics , Retroviridae/genetics
11.
Gene Ther ; 1(2): 136-8, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7584069

ABSTRACT

A set of retroviral vectors is described whose capacity for high efficiency transduction of functional genes into undifferentiated murine embryonic and haematopoietic cells makes them ideally suited for preclinical studies with murine models. Multiple unique cloning sites permit insertion of genes into the vectors such that no selectable marker exists or either the neomycin phosphotransferase (neo) gene, the hygromycin B phosphotransferase (hph) gene or the puromycin N-acetyl transferase (pac) gene is included as a dominantly acting selectable marker. Because the sequences in the viral gag region shown to improve the encapsidation of viral RNA have been modified to prevent viral protein synthesis and all env sequences have been removed to eliminate helper virus production by homologous recombination with packaging DNA, these vectors might prove useful in human gene therapy protocols.


Subject(s)
Genetic Therapy , Genetic Vectors , Retroviridae/genetics , Acetyltransferases/genetics , Animals , Base Sequence , Cells, Cultured , DNA, Recombinant/genetics , Gene Deletion , Genes, env , Genetic Markers , Humans , Kanamycin Kinase , Mice , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics
12.
Oncogene ; 9(1): 1-12, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7905617

ABSTRACT

The t(10;14) chromosomal translocation of T-cell acute lymphoblastic leukemia joins the T-cell receptor delta gene to a region upstream of a diverged homeobox-containing gene called HOX11. To better understand the pathogenetic role of HOX11 in leukemogenesis, post 5-fluorouracil-treated murine bone marrow cells were infected with a replication-defective retrovirus bearing the gene. Constitutive expression of HOX11 in hematopoietic precursors yielded cell lines at high frequency consisting of immature cells belonging to the myeloid lineage. HOX11-transformed cell lines displayed a strict dependence on IL-3 for their survival and proliferation in culture and were not leukemogenic. The results support the hypothesis that the transforming capacity of HOX11 derives from its ability to alter the expression of genes regulating hematopoietic differentiation and that secondary mutations promoting cell survival or stimulating proliferation are required for progression to malignancy. The findings further suggest that the oncogenic activity of HOX11 might not be restricted to T-cell leukemias in humans.


Subject(s)
Cell Transformation, Neoplastic , Cell Transformation, Viral , Genes, Homeobox , Hematopoietic Stem Cells/pathology , Leukemia/etiology , Retroviridae/genetics , Animals , Cells, Cultured , Female , Gene Expression Regulation , Humans , Interleukin-3/pharmacology , Leukemia/genetics , Mice , Mice, Inbred BALB C , Proviruses/genetics
13.
J Exp Med ; 178(4): 1175-88, 1993 Oct 01.
Article in English | MEDLINE | ID: mdl-8104229

ABSTRACT

Post 5-fluorouracil-treated murine bone marrow cells infected with a recombinant retrovirus (murine stem cell virus-interleukin 11 [MSCV-IL-11]) bearing a human IL-11 gene were transplanted into lethally irradiated syngeneic mice. Analysis of proviral integration sites in DNA prepared from hematopoietic tissues and purified cell populations of long-term reconstituted primary and secondary recipients demonstrated polyclonal engraftment by multipotential stem cells. High levels (100-1,500 U/ml) of IL-11 were detected in the plasma of the MSCV-IL-11 mice. Systemic effects of chronic IL-11 exposure included loss of body fat, thymus atrophy, some alterations in plasma protein levels, frequent inflammation of the eyelids, and often a hyperactive state. A sustained rise in peripheral platelet levels (approximately 1.5-fold) was seen throughout the observation period (4-17 wk). No changes were observed in the total number of circulating leukocytes in the majority of the transplanted animals (including 10 primary and 18 secondary recipients) despite a > 20-fold elevation in myeloid progenitor cell content in the spleen. The exceptions were members of one transplant pedigree which presented with myeloid leukemia during the secondary transplant phase. A clonal origin of the disease was determined, with significant expansion of the MSCV-IL-11-marked clone having occurred in the spleen of the primary host. Culturing of leukemic spleen cells from a quaternary recipient led to the establishment of a permanent cell line (denoted PGMD1). IL-11-producing PGMD1 myeloid leukemic cells are dependent on IL-3 for continuous growth in vitro and they differentiate into granulocytes and macrophages in response to granulocyte/macrophage colony-stimulating factor. The inability of autogenously produced IL-11 to support autonomous growth of PGMD1 cells argues against a mechanism of transformation involving a classical autocrine loop.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/pathology , Interleukin-11/physiology , Leukemia, Myeloid/etiology , Animals , B-Lymphocytes/microbiology , Bone Marrow/microbiology , Bone Marrow/pathology , Bone Marrow Transplantation , Chimera , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/microbiology , Humans , Hyperplasia , Interleukin-11/genetics , Interleukin-11/metabolism , Interleukin-6/physiology , Leukemia, Myeloid/pathology , Mice , Mice, Inbred BALB C , Platelet Count , Retroviridae/genetics
14.
Clin Exp Metastasis ; 11(2): 213-26, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8095198

ABSTRACT

We previously reported that a derivative of the interleukin-6 (IL-6)-dependent B9 B-cell hybridoma (B9/LPNU1L) constitutively expressing an interleukin-1 alpha (IL-1 alpha) gene introduced by retrovirus-mediated gene transfer preferentially metastasized to bone marrow following intravenous injection into unirradiated syngeneic BALB/c mice. B9/LPNU1L cells recovered from the femoral marrow of a recipient with hind limb paralysis (denoted B9/BM1) retained their IL-6-dependency yet displayed enhanced metastatic capacity during serial transplantation in vivo. In contrast, autonomously-growing B9 variants spontaneously arising in vitro or IL-6-independent B9 derivatives created by infection with recombinant IL-6 retroviruses rarely gave rise to experimental metastases in syngeneic BALB/c or nude mice. Examination of cell adhesion molecule profiles by immunofluorescence flow cytometry has revealed high levels of CD44, moderate levels of VLA-4 and low levels of LFA-1 on all B9-series cells. By comparison, ICAM-1 expression was significantly elevated on B9/BM1 cells, with independent isolates stably expressing about 4-fold higher levels which were paralleled by corresponding increases in the steady-state levels of ICAM-1 mRNA. L-Selectin was not expressed by any of the cell lines. Despite higher ICAM-1 levels, cell aggregation assays revealed that LFA-1-ICAM-1 adhesive interactions were not involved in the homotypic adhesion of B9/BM1 cells but rather that binding of CD44 to endogenously-synthesized hyaluronan was responsible. Furthermore, B9/BM1 cells expressing high levels of ICAM-1 were found to be less susceptible to cytolysis by natural killer (NK) cells than their weakly metastatic or nonmetastatic counterparts.


Subject(s)
Antigens, Neoplasm/physiology , B-Lymphocytes/immunology , Bone Marrow Cells , Bone Marrow/pathology , Cell Adhesion Molecules/physiology , Hybridomas/immunology , Hybridomas/pathology , Animals , Antibodies/pharmacology , B-Lymphocytes/pathology , Blotting, Northern , Cell Aggregation/immunology , Cell Aggregation/physiology , Cytotoxicity, Immunologic , Female , H-2 Antigens/physiology , Hybridomas/metabolism , Immunity, Cellular , Intercellular Adhesion Molecule-1 , Interleukin-1/biosynthesis , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplastic Cells, Circulating/immunology , Neoplastic Cells, Circulating/pathology , RNA, Messenger/genetics , Receptors, Lymphocyte Homing/immunology
15.
J Exp Med ; 176(4): 1149-63, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1402659

ABSTRACT

Lethally irradiated mice transplanted with bone marrow cells infected with a novel recombinant retrovirus (murine stem cell virus-interleukin 6 [MSCV-IL-6]) bearing a mouse IL-6 gene developed a fatal myeloproliferative disease within 4 wk of engraftment. The hematologic manifestations of the syndrome included elevated peripheral leukocyte counts (up to 430 x 10(3) cells/mm3) with a predominance of neutrophilic granulocytes, microcytic anemia, and thrombocytosis or thrombocytopenia. The mice showed extensive neutrophil infiltration of the lungs, liver, and occasionally lymph nodes, plus splenomegaly resulting from enhanced splenic myelopoiesis (30-60-fold increase in progenitor numbers). Despite the chronic stimulation of neutrophil excess by IL-6, bone marrow from affected mice was capable of repopulating the hematopoietic tissues (bone marrow and spleen) of lethally irradiated hosts during repeated serial transplantation. In the longest documented case, the progeny of a single MSCV-IL-6-marked cell transferred the myeloproliferative disease to two secondary, four tertiary, and two quaternary recipients (the clone endured for a total of 72 wk). These results, demonstrating considerable proliferative longevity of the IL-6-producing cells, support an in vivo role of IL-6 in the maintenance of hematopoietic precursors. Dysregulated IL-6 production also had significant systemic effects. The mice displayed increased mesangial cell proliferation in the kidney, frequent liver abnormalities, and marked alterations in plasma protein levels. Unlike previous studies where constitutive expression of exogenous IL-6 genes resulted in lymphoproliferative disorders characterized by massive plasmacytosis, minimal plasma cell expansion occurred in the MSCV-IL-6 mice during the observation period. Potential explanations for the differences in disease phenotypes observed in the present and previous studies are different cell types expressing the exogenous IL-6 genes, higher sustained circulating levels of IL-6 achieved using the MSCV-IL-6 retroviral delivery system, and/or the premature death (3-15 wk after transplantation) of the MSCV-IL-6 mice before the onset of plasmacytosis. This animal model should prove useful for further investigation of the function of IL-6 in normal and abnormal hematopoiesis and in inflammatory responses.


Subject(s)
Interleukin-6/genetics , Myeloproliferative Disorders/microbiology , Retroviridae/genetics , Animals , Blood Proteins/isolation & purification , Bone Marrow Transplantation/methods , Cell Line , Cells, Cultured , DNA, Viral/genetics , DNA, Viral/isolation & purification , Female , Genetic Vectors , Hematopoietic Stem Cells/pathology , Interleukin-6/analysis , Leukocyte Count , Mice , Mice, Inbred BALB C , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/pathology , Proviruses/genetics , Retroviridae/pathogenicity , Time Factors , Transfection , Virus Integration
SELECTION OF CITATIONS
SEARCH DETAIL
...