Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Sci Rep ; 2: 636, 2012.
Article in English | MEDLINE | ID: mdl-22957140

ABSTRACT

Cold atmospheric plasma (CAP), a technology based on quasi-neutral ionized gas at low temperatures, is currently being evaluated as a new highly selective alternative addition to existing cancer therapies. Here, we present a first attempt to identify the mechanism of CAP action. CAP induced a robust ~2-fold G2/M increase in two different types of cancer cells with different degrees of tumorigenicity. We hypothesize that the increased sensitivity of cancer cells to CAP treatment is caused by differences in the distribution of cancer cells and normal cells within the cell cycle. The expression of γH2A.X (pSer139), an oxidative stress reporter indicating S-phase damage, is enhanced specifically within CAP treated cells in the S phase of the cell cycle. Together with a significant decrease in EdU-incorporation after CAP, these data suggest that tumorigenic cancer cells are more susceptible to CAP treatment.


Subject(s)
Antineoplastic Agents/pharmacology , G2 Phase Cell Cycle Checkpoints , Plasma Gases/pharmacology , Skin Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Movement , Cell Survival , Keratinocytes/physiology , Mice , Mice, Inbred BALB C
2.
Oncogene ; 26(28): 4115-23, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17213805

ABSTRACT

Aberrant expression of the human homeobox-containing proto-oncogene TLX1/HOX11 inhibits hematopoietic differentiation programs in a number of murine model systems. Here, we report the establishment of a murine erythroid progenitor cell line, iEBHX1S-4, developmentally arrested by regulatable TLX1 expression. Extinction of TLX1 expression released the iEBHX1S-4 differentiation block, allowing erythropoietin-dependent acquisition of erythroid markers and hemoglobin synthesis. Coordinated activation of erythroid transcriptional networks integrated by the acetyltransferase co-activator CREB-binding protein (CBP) was suggested by bioinformatic analysis of the upstream regulatory regions of several conditionally induced iEBHX1S-4 gene sets. In accord with this notion, CBP-associated acetylation of GATA-1, an essential regulator of erythroid differentiation, increased concomitantly with TLX1 downregulation. Coimmunoprecipitation experiments and glutathione-S-transferase pull-down assays revealed that TLX1 directly binds to CBP, and confocal laser microscopy demonstrated that the two proteins partially colocalize at intranuclear sites in iEBHX1S-4 cells. Notably, the distribution of CBP in conditionally blocked iEBHX1S-4 cells partially overlapped with chromatin marked by a repressive histone methylation pattern, and downregulation of TLX1 coincided with exit of CBP from these heterochromatic regions. Thus, we propose that TLX1-mediated differentiation arrest may be achieved in part through a mechanism that involves redirection of CBP and/or its sequestration in repressive chromatin domains.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Homeodomain Proteins/physiology , Proto-Oncogene Proteins/physiology , Acetylation , Animals , Mice , Proto-Oncogene Mas , Up-Regulation
3.
Leuk Lymphoma ; 44(10): 1775-84, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14692533

ABSTRACT

Increasing evidence suggests a role for immunologic vaccination and therapy in the management of minimal residual myeloma. We have previously demonstrated a synergistic effect of combining the Th1 stimulating cytokine IL-12 with the co-stimulatory molecule CD80 in murine myeloma vaccination therapy. We reasoned that the efficacy of such treatment might be further improved by incorporating additional gene products which enhance the function of antigen presenting cells. Studies were therefore conducted with murine myeloma BM1 cells expressing Flt3L (membrane bound or soluble forms) or GM-CSF and the IL-12 x CD80 combination. Single agent and combined therapeutic approaches were explored. All gene-modified BM1 cells, except BM1/IL-12 x CD80, developed tumors when subcutaneously injected into BALB/c mice. As prophylactic tumor vaccines, the combined use of gene-modified BM1/sFlt3L+GM-CSF+IL-12 x CD80 was most effective, providing 100% protection against subsequent parental BM1 tumor challenge. By comparison, only partial protection was observed with any single gene-engineered tumor vaccine. Notably, IL-12 x CD80 coexpressing BM1 cell vaccines were the most effective therapeutic vaccine in a minimal disease model. Such protective vaccination was achieved by stimulation of lymphocyte proliferation and enhancement of cytotoxic lymphocyte activity.


Subject(s)
Cancer Vaccines/therapeutic use , Genetic Therapy , Immunotherapy , Multiple Myeloma/prevention & control , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/therapeutic use , Animals , B7-1 Antigen/metabolism , B7-1 Antigen/therapeutic use , Cancer Vaccines/metabolism , Cell Division , Combined Modality Therapy , Cytotoxicity, Immunologic , Female , Gene Transfer Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Interleukin-12/metabolism , Interleukin-12/therapeutic use , Ligands , Membrane Proteins/metabolism , Membrane Proteins/therapeutic use , Mice , Mice, Inbred BALB C , Mice, SCID , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Retroviridae/genetics , Spleen/immunology , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccination
4.
Cancer Gene Ther ; 8(5): 361-70, 2001 May.
Article in English | MEDLINE | ID: mdl-11477456

ABSTRACT

Synergy between interleukin-12 (IL-12) and B7-1 (CD80) for cancer immunotherapy has previously been demonstrated in animal models of breast cancer, lymphoma, and multiple myeloma. With a view to human clinical application, tricistronic retroviral and adenovirus vectors co-expressing IL-12 (IL-12p40 plus IL-12p35) and CD80 were constructed by utilizing two internal ribosome entry site (IRES) sequences to link the three cDNAs. A murine stem cell virus (MSCV)-based retroviral vector (MSCV-hIL12.B7) utilized distinct IRES sequences from the encephalomyocarditis virus (EMCV) and the foot-and-mouth disease virus (FMCV), whereas Ad5-based adenovirus vectors contained transcriptional units with two EMCV IRES sequences under the control of murine (AdMh12.B7) or human (AdHh12.B7) cytomegalovirus promoters. AdMh12.B7 was found to consistently direct higher levels of IL-12 and CD80 expression than AdHh12.B7 following infection of a number of human tumor cell lines. In preclinical studies, the human myeloma cell line U266 was infected with MSCV-hIL12.B7 and a resulting clonal cell line, U/MSCV-h12.B7, was generated with stable expression of CD80 and secreting IL-12 at 1 ng/24 h/10(6) cells. By comparison, following AdMh12.B7 infection, 81% of infected U266 cells (U/AdMh12.B7) expressed CD80 and secreted IL-12 at 25-50 ng/24 h/10(6) cells. Both engineered myeloma cell lines stimulated enhanced allogeneic mixed lymphocyte proliferation and provoked increases in cytotoxic T-lymphocyte responses and gamma-interferon release from normal donor lymphocytes exposed to parental U266 cells. These results suggest potential clinical utility of AdMh12.B7 in immunotherapy strategies for the treatment of multiple myeloma and other cancers.


Subject(s)
Adenoviridae/genetics , B7-1 Antigen/genetics , Genetic Vectors , Immunotherapy/methods , Interleukin-12/genetics , Neoplasms/therapy , Retroviridae/genetics , Tumor Cells, Cultured/drug effects , B7-1 Antigen/metabolism , Cytotoxicity Tests, Immunologic , Cytotoxicity, Immunologic , DNA, Complementary , Drug Therapy, Combination , Flow Cytometry , Gene Transfer Techniques , Humans , Interleukin-12/metabolism , Neoplasms/metabolism , Neoplasms/virology , T-Lymphocytes/immunology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/virology
5.
Biotechniques ; 30(5): 1028-34, 2001 May.
Article in English | MEDLINE | ID: mdl-11355338

ABSTRACT

Flow cytometric procedures are described to detect a "humanized" version of a new red fluorescent protein (DsRed) from the coral Discosoma sp. in conjunction with various combinations of three Aequorea victoria green fluorescent protein (GFP) variants--EYFP, EGFP, and ECFP. In spite of overlapping emission spectra, the combination of DsRed with EYFP, EGFP, and ECFP generated fluorescence signals that could be electronically compensated in real time using dual-laser excitation at 458 and 568 nm. Resolution of fluorescence signals from DsRed, EYFP, and EGFP was also readily achieved by single-laser excitation at 488 nm. Since many flow cytometers are equipped with an argon-ion laser that can be tuned to 488 nm, the DsRed/EYFP/EGFP combination is expected to have broad utility for facile monitoring of gene transfer and expression in mammalian cells. The dual-laser technique is applicable for use on flow cytometers equipped with tunable multiline argon-ion and krypton-ion lasers, providing the framework for studies requiring simultaneous analysis of four fluorescent gene products within living cells.


Subject(s)
Bacterial Proteins/analysis , Flow Cytometry , Luminescent Proteins/analysis , Retroviridae/genetics , 3T3 Cells , Animals , Bacterial Proteins/genetics , Flow Cytometry/methods , Gene Expression , Genetic Variation , Genetic Vectors , Green Fluorescent Proteins , Lasers , Luminescent Proteins/genetics , Mice , Microscopy, Fluorescence , Spectrometry, Fluorescence , Transfection , Red Fluorescent Protein
6.
Blood ; 97(8): 2413-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290605

ABSTRACT

Translocations involving fibroblast growth factor receptor 3 (fgfr3) have been identified in about 25% of patients with myeloma. To directly examine the oncogenic potential of fgfr3, murine bone marrow (BM) cells were transduced with retroviral vectors containing either wild-type fgfr3 or an activated mutant form of the receptor, fgfr3-TD. Mice transplanted with FGFR3-TD-expressing BM developed a marked leukocytosis and lethal hematopoietic cell infiltration of multiple tissues within 6 weeks of transplantation. Secondary and tertiary recipients of spleen or BM from primary fgfr3-TD mice also developed tumors within 6 to 8 weeks. Analysis of the circulating tumor cells revealed a pre-B-cell phenotype in most mice, although immature T-lymphoid or mature myeloid populations also predominated in some animals. Enhanced lymphoid but not myeloid colony formation was observed in the early posttransplantation period and only interleukin 7 and FGF-responsive pre-B-cell lines could be established from tumors. Cell expansions in primary recipients appeared polyclonal, whereas tumors in later passages exhibited either clonal B- or T-cell receptor gene rearrangements. Mice transplanted with wild-type FGFR3-expressing BM developed delayed pro-B-cell lymphoma/leukemias approximately 1 year after transplantation. These studies confirm that FGFR3 is transforming and can produce lymphoid malignancies in mice.


Subject(s)
Cell Transformation, Neoplastic/genetics , Hematopoietic Stem Cells/cytology , Lymphoproliferative Disorders/etiology , Multiple Myeloma/genetics , Neoplasm Proteins/physiology , Oncogenes , Protein-Tyrosine Kinases , Animals , Bone Marrow Transplantation , Clone Cells/pathology , Female , Fibroblast Growth Factors/pharmacology , Gene Rearrangement, B-Lymphocyte , Gene Rearrangement, T-Lymphocyte , Humans , Interleukin-7/pharmacology , Leukocytosis/etiology , Leukocytosis/genetics , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/genetics , Lymphoproliferative Disorders/genetics , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Radiation Chimera , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/physiology , Recombinant Fusion Proteins/physiology , Spleen/transplantation
7.
Stem Cells ; 19(2): 118-24, 2001.
Article in English | MEDLINE | ID: mdl-11239166

ABSTRACT

Hematologic diseases potentially benefiting from gene-based therapies involving hematopoietic stem cells (HSCs) include hereditary hemoglobinopathies, immunodeficiency syndromes, and congenital bleeding disorders such as hemophilia A, as well as acquired diseases like AIDS. Successful treatment of these blood diseases with gene-modified HSCs requires high efficiency gene delivery to the target cell population and persistence of transgene expression following differentiation. We review flow cytometric procedures that permit simultaneous, noninvasive measurements of transgene expression and phenotypic discrimination of hematopoietic cell subsets. Central to this approach has been the recent development of a spectrum of blue, cyan, and yellowish-green fluorescent reporters based on the jellyfish Aequorea victoria green fluorescent protein and the discovery of a red fluorescent protein in DISCOSOMA: coral. This methodology should facilitate the optimization of oncoretroviral and lentiviral vectorology and HSC transduction protocols for the ultimate purpose of HSC-directed gene therapy.


Subject(s)
Genes, Reporter , Hematopoietic Stem Cells/cytology , Luminescent Proteins , Animals , Cell Lineage , Drug Resistance , Green Fluorescent Proteins , Hematopoietic Stem Cells/classification , Humans , Luminescent Proteins/genetics , Membrane Proteins
8.
Mol Ther ; 2(5): 458-69, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11082319

ABSTRACT

Accumulated data indicate that current generation lentiviral vectors, which generally utilize an internal human cytomegalovirus (CMV) immediate early region enhancer-promoter to transcribe the gene of interest, are not yet optimized for efficient expression in human hematopoietic stem/progenitor cells (HSPCs). As a first step toward this goal, we constructed self-inactivating derivatives of the HIV-1-based transfer vector pHR' containing the enhanced green fluorescent protein (GFP) gene as reporter and the Woodchuck hepatitis virus posttranscriptional regulatory element (WPRE). GFP expression was driven by a variety of strong viral and cellular promoters, including the murine stem cell virus (MSCV) long terminal repeat (LTR), a Gibbon ape leukemia virus (GALV) LTR, the human elongation factor 1alpha (EF1alpha) promoter, the composite CAG promoter (consisting of the CMV immediate early enhancer and the chicken beta-actin promoter), and the human phosphoglycerate kinase 1 (PGK) promoter. In contrast to results obtained in human embryonic kidney 293T cells and fibrosarcoma HT1080 cells, in which the CMV promoter expressed GFP at the highest levels, significantly higher levels of GFP expression (3- to 5-fold) were achieved with the MSCV LTR, the EF1alpha promoter, and the CAG promoter in the human HSPC line KG1a. Removal of the WPRE indicated that it stimulated GFP expression from all of the vectors in KG1a cells (up to 3-fold), although it only marginally improved the performance of the intron-containing EF1alpha and CAG promoters (<1.5-fold stimulation). The vectors using the MSCV LTR, the GALV LTR, and the PGK promoter were the most efficient at transducing primary human CD34+ cord blood progenitors under the conditions employed. High-level GFP expression in the NOD/SCID xenograft model was demonstrated with the pHR' derivative bearing the MSCV LTR. These new lentiviral vector backbones provide a basis for the rational design of improved delivery vehicles for human HSPC gene transfer applications.


Subject(s)
Gene Expression , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Antigens, CD34/analysis , Cell Line , Fetal Blood/metabolism , Genetic Vectors/metabolism , Green Fluorescent Proteins , HIV-1/genetics , Humans , Indicators and Reagents/metabolism , Lentivirus/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Promoter Regions, Genetic , Transduction, Genetic , Transgenes , Tumor Cells, Cultured
9.
Leuk Lymphoma ; 39(3-4): 241-56, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11342305

ABSTRACT

The mammalian PBX and Meis proteins belong to the TALE (three-amino acid-loop-extension) superfamily of homeodomain-containing transcription factors. Members of both the PBX and Meis groups have been implicated in tumorigenesis and are known to cooperatively bind DNA with Class I (clustered) HOX homeoproteins. Here we show that PBX and Meis homeoproteins cooperatively bind the PBX-responsive sequence in vitro with the oncoprotein encoded by the non-clustered homeobox gene HOX11 activated by the t(10;14)(q24;q11) chromosomal translocation in T-cell acute lymphoblastic leukemia (T-ALL). An FPWME motif N-terminal to the homeodomain is required for interaction with PBX proteins, which appears to confer DNA-binding specificity to HOX11. PBX proteins are highly expressed in HOX11 immortalized/transformed hematopoietic cells; in particular, the 10q24 translocation-carrying T-ALL Sil and K3P lines were found to selectively express PBX2. Ectopic retroviral-directed overexpression of PBX2 in concert with HOX11 in NIH3T3 cells resulted in decreased contact inhibition of growth as evidenced by focus formation in confluent cell monolayers. The accumulated data are thus consistent with a role of TALE homeoproteins in HOX11-mediated leukemogenesis.


Subject(s)
Homeodomain Proteins/genetics , Leukemia, T-Cell/genetics , Oncogene Proteins/genetics , Cell Transformation, Neoplastic/genetics , Genes, Homeobox , Humans , Leukemia, T-Cell/etiology , Proto-Oncogene Proteins
10.
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
11.
Int J Oncol ; 15(1): 173-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10375612

ABSTRACT

Interleukin-6 (IL-6) is implicated in the in vivo proliferation of malignant plasma cells in multiple myeloma. To define the molecular basis of the IL-6-induced mitogenic response in myeloma cells, we applied STAR (subtractive transcriptional amplification of mRNA), a new differential expression analysis technology, to isolate mRNAs preferentially expressed in IL-6-treated versus untreated cultures of the factor-responsive myeloma cell line U266. From the resulting collection of STAR clones, sequence information was obtained for a total of 72 distinct transcripts. Of these, 29 were found to correspond to known genes, 22 matched expressed sequence tags in public databases and 21 showed no sequence similarity to any existing entries. Among the known genes uncovered in the screen were those encoding proteins that function in cell division, cell signalling and gene/protein expression. Northern blot analysis documented that two transcription factor genes chosen for further study, c-myc promoter-binding protein (MBP-1) and X-box binding protein 1 (XBP-1), were up-regulated in U266 cells about 3-fold relative to the cell cycle-dependent beta-actin gene 12 h after IL-6 treatment. Both genes were also similarly up-regulated by IL-6 in factor-dependent ANBL-6 myeloma cells. These results indicate that MBP-1 and XBP-1 are IL-6 genes in myeloma cells; as such, they may play a role in IL-6-mediated growth control in multiple myeloma.


Subject(s)
DNA-Binding Proteins/genetics , Interleukin-6/pharmacology , Multiple Myeloma/genetics , Neoplasm Proteins/genetics , Phosphopyruvate Hydratase , Transcription Factors/genetics , Tumor Suppressor Proteins , Base Sequence , Biomarkers, Tumor , Cell Division/drug effects , DNA, Complementary/genetics , Expressed Sequence Tags , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/physiology , Molecular Sequence Data , Multiple Myeloma/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/isolation & purification , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Regulatory Factor X Transcription Factors , Signal Transduction/drug effects , Subtraction Technique , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , X-Box Binding Protein 1
12.
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
13.
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
14.
Mol Cell Biol ; 18(3): 1525-33, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9488469

ABSTRACT

Signals propagated via the gp130 subunit of the interleukin-6 (IL-6)-type cytokine receptors mediate, among various cellular responses, proliferation of hematopoietic cells and induction of acute-phase plasma protein (APP) genes in hepatic cells. Hematopoietic growth control by gp130 is critically dependent on activation of both STAT3 and protein tyrosine phosphatase 2 (SHP-2). To investigate whether induction of APP genes has a similar requirement for SHP-2, we constructed two chimeric receptors, G-gp130 and G-gp130(Y2F), consisting of the transmembrane and cytoplasmic domains of gp130 harboring either a wild-type or a mutated SHP-2 binding site, respectively, fused to the extracellular domain of the granulocyte colony-stimulating factor (G-CSF) receptor. Rat hepatoma H-35 cells stably expressing the chimeric receptors were generated by retroviral transduction. Both chimeric receptors transmitted a G-CSF-induced signal characteristic of that triggered by IL-6 through the endogenous gp130 receptor; i.e., both activated the appropriate JAK, induced DNA binding activity by STAT1 and STAT3, and up-regulated expression of the target APP genes, those for alpha-fibrinogen and haptoglobin. Notwithstanding these similarities in the patterns of signaling responses elicited, mutation of the SHP-2 interaction site in G-gp130(Y2F) abrogated ligand-activated receptor recruitment of SHP-2 as expected. Moreover, the tyrosine phosphorylation state of the chimeric receptor, the associated JAK activity, and the induced DNA binding activity of STAT1 and STAT3 were maintained at elevated levels and for an extended period of time in G-gp130(Y2F)-expressing cells following G-CSF treatment compared to that in cells displaying the G-gp130 receptor. H-35 cells ectopically expressing G-gp130(Y2F) were also found to display an enhanced sensitivity to G-CSF and a higher level of induction of APP genes. Overexpression of the enzymatically inactive SHP-2 enhanced the signaling by the wild-type but not by the Y2F mutant G-gp130 receptor. These results indicate that gp130 signaling for APP gene induction in hepatic cells differs qualitatively from that controlling the proliferative response in hematopoietic cells in not being strictly dependent on SHP-2. The data further suggest that SHP-2 functions normally to attenuate gp130-mediated signaling in hepatic (and, perhaps, other) cells by moderating JAK action.


Subject(s)
Acute-Phase Proteins/biosynthesis , Antigens, CD/metabolism , Membrane Glycoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins , Signal Transduction , Acute-Phase Proteins/genetics , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cytokine Receptor gp130 , DNA-Binding Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Janus Kinase 2 , Liver/cytology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Protein Phosphatase 2 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Rats , STAT1 Transcription Factor , STAT3 Transcription Factor , TYK2 Kinase , Trans-Activators/metabolism , Tumor Cells, Cultured
15.
Hematology ; 3(5): 365-74, 1998.
Article in English | MEDLINE | ID: mdl-27414080

ABSTRACT

There has been little improvement in the treatment of multiple myeloma over the past 25 years. Disease inevitably reoccurs in patients who receive chemotherapy of melphalan and prednisone or combinations of alkylating agents. Autologous hematopoietic stem cell transplantation can increase remission rates and prolong diseasefree and overall survival. However, all transplanted myeloma patients ultimately relapse. The ineffectiveness of conventional induction and maintenance therapies in multiple myeloma has motivated the search for alternative treatment strategies. Immunotherapy involving cancer vaccines is one such alternative where the intent is to induce a host antitumour immune response. In this study, we employed a syngeneic murine model of multiple myeloma developed in our laboratory to examine the consequence of combined expression of interleukin-12 (IL-12) and the B7-1 costimulatory molecule on myeloma immuno genicity. We show that the IL-12/B7-1 immunogene combination was efficacious in evoking systemic protective immunity against unmodified parental myeloma cells. These findings suggest that autologous myeloma cells engineered to co-express IL-12 and B7-1 may hold promise as cancer vaccines for consolidation therapy in multiple myeloma.

16.
J Biol Chem ; 272(26): 16216-23, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9195922

ABSTRACT

Leptin, an adipocyte-secreted hormone, is one of the central regulators of body weight homeostasis. In humans and rodents, two major forms of leptin receptors (OB-R) are expressed. The short form (OB-RS), considered to lack signaling capability, is detected in many organs. In contrast, OB-R long form (OB-RL) predominates in the hypothalamus, but is also present at low levels in peripheral tissues. Transient transfection experiments have demonstrated that OB-RL transduces an intracellular signaling similar to interleukin (IL)-6 type-cytokine receptors. To define the specificity by which OB-R induces genes and cooperates with signal transduction pathways utilized by other hormones and cytokines, rat and human hepatoma cell lines were generated which stably express human OB-RL. Hepatoma cell lines selected for appreciable levels of OB-RL mRNA display enhanced leptin binding and responded to leptin with an IL-6 receptor-like signaling that includes the activation of STAT proteins, induction of acute-phase plasma proteins, and synergism with IL-1 and tumor necrosis factor-alpha. A leptin-mediated recruitment of phosphatidylinositol 3-kinase to insulin receptor substrate-2 was also detected. However, no significant tyrosine phosphorylation of insulin receptor substrate-2 and modulation of the immediate cell response to insulin were observed. The data suggest that OB-RL action in hepatic cells is equivalent to that of IL-6 receptor. However, leptin does not play a specific role in muting insulin action on hepatoma cells and therefore may not contribute to the diabetic symptoms associated with obesity.


Subject(s)
Antigens, CD/physiology , Carrier Proteins/physiology , Interleukin-6 , Liver/metabolism , Receptors, Cell Surface , Receptors, Interleukin/physiology , Acute-Phase Proteins/genetics , Animals , Carrier Proteins/genetics , DNA-Binding Proteins/physiology , Growth Inhibitors/pharmacology , Humans , Insulin , Leptin , Leukemia Inhibitory Factor , Lymphokines/pharmacology , Mice , Phosphatidylinositol 3-Kinases , Phosphotransferases (Alcohol Group Acceptor)/drug effects , Proteins/pharmacology , RNA, Messenger/analysis , Rats , Receptors, Interleukin-6 , Receptors, Leptin , STAT1 Transcription Factor , STAT3 Transcription Factor , Trans-Activators/physiology , Tumor Cells, Cultured
17.
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
18.
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
19.
Hum Gene Ther ; 8(18): 2217-28, 1997 Dec 10.
Article in English | MEDLINE | ID: mdl-9449375

ABSTRACT

The A20 murine B cell lymphoma was transfected with B7-1 and subsequently these variants and vector control variants were retrovirally infected to express murine interleukin-12 (mIL-12). In vitro data showed that the B7-1 variants enhanced secretion of IL-2 and IL-4 by allogeneic T cells in mixed lymphocyte tumor cultures. While IL-12 variants stimulated IFN-gamma, variants expressing both B7-1 and IL-12 stimulated IFN-gamma, IL-2, and IL-4 secretion. Tumorigenicity experiments showed that whereas B7-1 delayed tumor onset, only the mIL-12 variants with or without B7-1 were completely rejected in syngeneic hosts. In addition, tumor-free mice were protected against subsequent challenge with the parental unmodified cells and had enhanced cytotoxic T lymphocyte (CTL) lysis activity. Results from minimal disease mixing experiments demonstrated that only the A20/B7-1/mIL-12 variant was able to reject A20 unmodified cells inoculated at the same site, whereas prolonged survival was observed when the A20 parental cells were inoculated at different sites. Depletion studies and injections into nu-/nu- mice demonstrated that both CD4+ and CD8+ T cells may mediate immunity. These data suggest that vaccinations with tumor cells genetically modified to express both B7-1 and IL-12 may alter cytokine profiles and generate CTL activity and, thus, the mechanisms of enhanced antitumor immunity may be multifactorial.


Subject(s)
B7-1 Antigen/genetics , Interleukin-12/genetics , Lymphoma, B-Cell/immunology , Neoplasms, Experimental/immunology , Animals , B7-1 Antigen/biosynthesis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Female , Genetic Engineering , Lymphoma, B-Cell/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms, Experimental/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...