Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 801
Filter
1.
Blood ; 136(18): 2003-2017, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32911536

ABSTRACT

The majority of childhood leukemias are precursor B-cell acute lymphoblastic leukemias (pB-ALLs) caused by a combination of prenatal genetic predispositions and oncogenic events occurring after birth. Although genetic predispositions are frequent in children (>1% to 5%), fewer than 1% of genetically predisposed carriers will develop pB-ALL. Although infectious stimuli are believed to play a major role in leukemogenesis, the critical determinants are not well defined. Here, by using murine models of pB-ALL, we show that microbiome disturbances incurred by antibiotic treatment early in life were sufficient to induce leukemia in genetically predisposed mice, even in the absence of infectious stimuli and independent of T cells. By using V4 and full-length 16S ribosomal RNA sequencing of a series of fecal samples, we found that genetic predisposition to pB-ALL (Pax5 heterozygosity or ETV6-RUNX1 fusion) shaped a distinct gut microbiome. Machine learning accurately (96.8%) predicted genetic predisposition using 40 of 3983 amplicon sequence variants as proxies for bacterial species. Transplantation of either wild-type (WT) or Pax5+/- hematopoietic bone marrow cells into WT recipient mice revealed that the microbiome is shaped and determined in a donor genotype-specific manner. Gas chromatography-mass spectrometry (GC-MS) analyses of sera from WT and Pax5+/- mice demonstrated the presence of a genotype-specific distinct metabolomic profile. Taken together, our data indicate that it is a lack of commensal microbiota rather than the presence of specific bacteria that promotes leukemia in genetically predisposed mice. Future large-scale longitudinal studies are required to determine whether targeted microbiome modification in children predisposed to pB-ALL could become a successful prevention strategy.


Subject(s)
Disease Susceptibility , Dysbiosis/complications , Feces/microbiology , Gastrointestinal Microbiome , Leukemia, Experimental/prevention & control , PAX5 Transcription Factor/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Animals , Female , Leukemia, Experimental/genetics , Leukemia, Experimental/microbiology , Leukemia, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/microbiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
2.
J Immunol ; 184(6): 3072-8, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20154210

ABSTRACT

Chlamydophila pneumoniae infection of the vascular wall as well as activation of the transcription factor IFN regulatory factor (IRF)3 have been linked to development of chronic vascular lesions and atherosclerosis. The innate immune system detects invading pathogens by use of pattern recognition receptors, some of which are able to stimulate IRF3/7 activation and subsequent type I IFN production (e. g., IFN-beta). In this study, we show that infection of human endothelial cells with C. pneumoniae-induced production of IFN-beta, a cytokine that so far has been mainly associated with antiviral immunity. Moreover, C. pneumoniae infection led to IRF3 and IRF7 nuclear translocation in HUVECs and RNA interference experiments showed that IRF3 and IRF7 as well as the mitochondrial antiviral signaling (MAVS) were essential for IFN-beta induction. Finally, C. pneumoniae replication was enhanced in endothelial cells in which IRF3, IRF7, or MAVS expression was inhibited by small interfering RNA and attenuated by IFN-beta treatment. In conclusion, C. pneumoniae infection of endothelial cells activates an MAVS-, IRF3-, and IRF7-dependent signaling, which controls bacterial growth and might modulate development of vascular lesions.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Chlamydophila pneumoniae/growth & development , Chlamydophila pneumoniae/immunology , Endothelium, Vascular/immunology , Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Interferon-beta/physiology , Mitochondrial Proteins/physiology , RNA Interference/physiology , Cells, Cultured , Down-Regulation/immunology , Endothelium, Vascular/microbiology , Endothelium, Vascular/virology , Humans , Immunity, Innate , Interferon-beta/biosynthesis , Interferon-beta/genetics , Leukemia, Experimental/immunology , Leukemia, Experimental/microbiology , Leukemia, Experimental/virology , Moloney murine leukemia virus/immunology , RNA, Viral/antagonists & inhibitors , Retroviridae Infections/immunology , Retroviridae Infections/microbiology , Retroviridae Infections/virology , Signal Transduction/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/microbiology , Tumor Virus Infections/virology
3.
Oncogene ; 14(12): 1471-9, 1997 Mar 27.
Article in English | MEDLINE | ID: mdl-9136991

ABSTRACT

We have shown recently that a retrovirus vector expressing a natural mutant form of the PML-RAR alpha protein characteristic of human acute promyelocytic leukaemia can transform early chicken hematopoietic progenitors (Altabef et al., 1996). Neither truncated PML nor truncated RAR alpha alone could induce transformation which suggest that the two domains should cooperate for the oncogenicity of the fusion product. To further investigate the mechanisms of this co-operation, we have tested whether a truncated RAR alpha could cooperate with the v-erbB oncogene. This oncogene has previously been shown to co-operate with the rearranged thyroid hormone receptor, v-erbA, to transform erythrocytic progenitors. We show that v-erbB and a truncated RAR alpha co-operate when expressed simultaneously as independent products to transform very early chicken haematopoietic cells close to pluripotent stage. In addition, we show that v-erbB alters transcriptional abilities of RAR alpha by both enhancing its effects on RARE and reducing those on AP-1. Therefore, RAR alpha is able to co-operate with different kinds of proteins to induce transformation of early haematopoietic cells. This strongly suggests that RAR alpha are involved in the differentiation commitment of early haematopoietic progenitors during the normal process of haematopoietic differentiation. These data bring new insights in the mechanisms of oncogenic transformation by rearranged RAR alpha.


Subject(s)
Cell Transformation, Viral , Hematopoietic Stem Cells/cytology , Leukemia, Experimental/genetics , Oncogene Proteins v-erbB/physiology , Receptors, Retinoic Acid/physiology , Animals , Chick Embryo , Chickens , Gene Expression Regulation, Neoplastic , Leukemia, Experimental/microbiology , Leukemia, Experimental/pathology , Retinoic Acid Receptor alpha , Retroviridae , Sequence Deletion , Structure-Activity Relationship , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
4.
J Virol ; 68(8): 5174-83, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8035516

ABSTRACT

Recombinant murine leukemia viruses (MuLVs) from high-leukemia-incidence mouse strains typically acquire pathogenic U3 region sequences from the genome of the endogenous xenotropic virus, Bxv-1. However, a recombinant virus isolated from a leukemic HRS/J mouse and another from a CWD mouse contained U3 regions that lacked genetic markers of Bxv-1. The U3 regions of both recombinants were derived from the endogenous ecotropic virus Env-1 and had retained a single enhancer element. However, compared with that of Emv-1, the U3 region of each of the recombinant viruses contained five nucleotide substitutions, one of which was shared. To determine the biological significance of these substitutions, chimeric ecotropic viruses that contained the U3 region from one of the two recombinant viruses or from Emv-1 were injected into NIH Swiss mice. All three of the chimeric ecotropic viruses were leukemogenic following a long latency. Despite the presence of an enhancer core motif that is known to contribute to the leukemogenicity of the AKR MuLV SL3-3, the HRS/J virus U3 region induced lymphomas only slightly more rapidly than the allelic Emv-1 sequences. The chimeric virus with the U3 region of the CWD recombinant caused lymphomas more frequently and more rapidly than either of the other two viruses. The results support the hypothesis that one or more of the five nucleotide substitutions in the U3 regions of the recombinants contribute to viral pathogenicity. Comparison of DNA sequences suggests that the pathogenicity of the CWD virus U3 region was related to a sequence motif that is shared with Bxv-1 and is recognized by the basic helix-loop-helix class of transcription factors.


Subject(s)
Leukemia Virus, Murine/pathogenicity , 3T3 Cells , Animals , Base Sequence , Cell Line , DNA, Viral , Enhancer Elements, Genetic , Leukemia Virus, Murine/genetics , Leukemia, Experimental/microbiology , Leukemia, Experimental/pathology , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Proviruses/genetics , Rats , Recombination, Genetic , Retroviridae Infections/microbiology , Retroviridae Infections/pathology , Sequence Homology, Nucleic Acid , Tumor Virus Infections/microbiology , Tumor Virus Infections/pathology
5.
J Virol ; 68(8): 5194-203, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7518532

ABSTRACT

Polytropic murine leukemia viruses (MuLVs) arise in mice by recombination of ecotropic MuLVs with endogenous retroviral envelope genes and have been implicated in the induction of hematopoietic proliferative diseases. Inbred mouse strains contain many endogenous sequences which are homologous to the polytropic env genes; however, the extent to which particular sequences participate in the generation of the recombinants is unknown. Previous studies have established antigenic heterogeneity among the env genes of polytropic MuLVs, which may reflect recombination with distinct endogenous genes. In the present study, we have examined many polytropic MuLVs and found that nearly all isolates fall into two mutually exclusive antigenic subclasses on the basis of the ability of their SU proteins to react with one of two monoclonal antibodies, termed Hy 7 and MAb 516. Epitope-mapping studies revealed that reactivity to the two antibodies is dependent on the identity of a single amino acid residue encoded in a variable region of the receptor-binding domain of the env gene. This indicated that the two antigenic subclasses of MuLVs arose by recombination with distinct sets of endogenous genes. Evaluation of polytropic MuLVs in mice revealed distinctly different ratios of the two subclasses after inoculation of different ecotropic MuLVs, suggesting that individual ecotropic MuLVs preferentially recombine with distinct sets of endogenous polytropic env genes.


Subject(s)
Epitopes/immunology , Leukemia Virus, Murine/immunology , Leukemia, Experimental/microbiology , Retroviridae Infections/microbiology , Tumor Virus Infections/microbiology , 3T3 Cells , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Base Sequence , Cell Line , DNA, Viral , Leukemia Virus, Murine/classification , Mice , Mink , Molecular Sequence Data , Point Mutation , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
6.
Leukemia ; 8(7): 1202-13, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8035613

ABSTRACT

The mechanism by which non-oncogene-bearing, slowly transforming retroviruses induce leukemia is not well understood, but appears to represent a multi-step process. Cell lines have been isolated following in vitro infection of lymphoid cells with radiation leukemia virus (RadLV) and they have been used to develop a two-step model for leukemia development. Thymic tumors were induced when one of the cell lines, C1-V13D, was inoculated into CBA/H mouse thymus. Upon reisolation of C1-V13D cells after one, two and three passages through thymus, individual cloned cell lines displayed increased tumorigenic potential compared with the non-tumorigenic parental line. Southern analysis has been used to track any genetic changes occurring while cells undergo further transformation and become increasingly tumorigenic. Specifically, retrovirus integration has been monitored in clones derived from C1-V13D at the primary, secondary and tertiary passage through thymus using probes specific for long terminal repeat (LTR), gag, pol and env genes of RadLV. The data indicate multiple ecotropic retrovirus integration sites in C1-V13D cells. Primary thymic tumors also showed the integration of a new recombinant or defective virus. There was no evidence that new ecotropic retrovirus integration had occurred during subsequent passage of primary tumors through the thymus, i.e. during the progression to oncogenesis. All data indicate an important role for the thymic environment in the development of a fully transformed cell.


Subject(s)
Cell Transformation, Viral/radiation effects , Leukemia, Experimental/genetics , Leukemia, Experimental/microbiology , Oncogenes/radiation effects , Radiation Leukemia Virus/genetics , Animals , Cell Division/physiology , DNA, Viral/genetics , Disease Models, Animal , Female , Male , Mice , Mice, Inbred CBA , Thymus Gland/cytology , Thymus Gland/microbiology , Thymus Neoplasms/microbiology , Thymus Neoplasms/pathology , Virus Integration
7.
Cancer Lett ; 82(1): 89-94, 1994 Jul 15.
Article in English | MEDLINE | ID: mdl-8033074

ABSTRACT

The effects of tamoxifen (TAM), its 3-hydroxy congener droloxifene (DROL) and 17 beta-estradiol were investigated on leukemogenesis induced in BALB/c mice by Rauscher murine leukemia virus (RLV). Multiple applications of each compound, in a dose-dependent manner, resulted in reduced virus titer in the serum, delayed onset of splenomegaly and significant prolongation of survival. Although 17 beta-estradiol proved most effective, prevention of disease was not achieved either by short- or long-term treatment with any of the drugs tested.


Subject(s)
Estradiol/pharmacology , Leukemia, Experimental/microbiology , Rauscher Virus/drug effects , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Animals , Antineoplastic Agents , Estradiol/administration & dosage , Estrogen Antagonists , Female , Leukemia, Experimental/prevention & control , Male , Mice , Mice, Inbred BALB C , Tamoxifen/administration & dosage
8.
Virology ; 202(1): 500-5, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7516600

ABSTRACT

To study the possible role of immune selection in the in vivo generation of pathogenic recombinant murine leukemia viruses (MuLV), we have constructed recombinant vaccinia viruses (rVV) expressing the envelope genes of three MuLV: AKR623, MCF247, and MCF13. rVV expressing either AKR623 or MCF247 env could prime H-2b mice for anti-AKR/Gross virus CTL responses, and stimulate the in vitro generation of CTL from the spleens of mice immunized with an AKR/Gross virus-positive lymphoma. MC57 (H-2b) cells infected with either 623EnvVac or 247EnvVac could serve as targets for ARK/Gross virus-specific CTL. Cells infected with the rVV expressing MCF13 env, however, were lysed much less efficiently by these CTL. 13EnvVac was also ineffective in priming or stimulating retrovirus-specific CTL. Finally, experiments with synthetic peptides and minigenes suggested that the reduced immunogenicity of the MCF13 envelope protein likely resulted from a single amino acid substitution within an immunodominant epitope of the p15E (TM) protein. The region of MCF13 env that encodes this epitope is derived from an endogenous xenotropic virus, while the allelic sequences in MCF247 are of ecotropic virus origin. These results suggest the potential for recombination within the MuLV envelope gene to allow escape from host cellular immune responses.


Subject(s)
Leukemia, Experimental/immunology , Mink Cell Focus-Inducing Viruses/immunology , Retroviridae Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Virus Infections/immunology , Viral Envelope Proteins/immunology , Animals , Base Sequence , DNA, Viral , Epitopes/immunology , Female , H-2 Antigens , Immunization , Leukemia, Experimental/microbiology , Male , Mice , Mink Cell Focus-Inducing Viruses/pathogenicity , Molecular Sequence Data , Retroviridae Infections/microbiology , Tumor Virus Infections/microbiology
10.
J Virol ; 68(2): 1165-72, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8289345

ABSTRACT

We have investigated the phenotypic changes that take place during the process of neoplastic transformation in the thymocytes of C57BL/Ka mice infected by the radiation leukemia virus (RadLV). By the combined use of antibodies against the envelope glycoprotein gp70 of RadLV, the transformation-associated cell surface marker 1C11, and the CD3-T-cell receptor (TCR) complex, we found that in the RadLV-infected thymus, the earliest expression of viral gp70 is in 1C11hi cells; a small but significant percentage of these cells also express CD3. A first wave of viral replication, manifested by the expression of high levels of gp70 in thymocytes (over 70% positive), reaches a peak at 2 weeks; during this period, no significant changes are observed in the expression of 1C11 or CD3. The population of gp70+ cells is drastically reduced at 3 to 4 weeks after infection. However, a second cohort of gp70+ cells appears after 4 weeks, and these cells express high levels of 1C11 and TCR determinants as well. RadLV-induced lymphomas differ from normal thymocytes in their CD4 CD8 phenotype, with domination by one or more subsets. Characterization of TCR gene rearrangements in RadLV-induced lymphomas shows that most of these tumors are clonal or oligoclonal with respect to the J beta 2 TCR gene, while the J beta 1 TCR gene is rearranged in a minority (4 of 11) of lymphomas. TCR V beta repertoire analysis of 12 tumors reveals that 6 (50%) express exclusively the V beta 6 gene product, 2 (17%) are V beta 5+, and 1 (8%) each are V beta 8+ and V beta 9+. In normal C57BL/Ka mice, V beta 6 is expressed on 12%, V beta 5 is expressed on 9%, V beta 8 is expressed on 22%, and V beta 9 is expressed on 4% of TCRhi thymocytes. Thus, it appears that RadLV-induced thymic lymphomas are not randomly selected with respect to expressed TCR V beta type.


Subject(s)
Lymphoma/immunology , Radiation Leukemia Virus/growth & development , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Retroviridae Infections/immunology , Thymus Neoplasms/immunology , Tumor Virus Infections/immunology , Animals , Antigens, Differentiation/analysis , CD3 Complex/analysis , CD4 Antigens/analysis , CD8 Antigens/analysis , Cell Transformation, Neoplastic , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Leukemia, Experimental/immunology , Leukemia, Experimental/microbiology , Lymphoma/microbiology , Mice , Mice, Inbred C57BL , Retroviridae Proteins, Oncogenic/analysis , Selection, Genetic , Thymus Gland/cytology , Thymus Neoplasms/microbiology , Viral Envelope Proteins/analysis , Virus Replication
11.
Folia Biol (Praha) ; 40(3): 113-8, 1994.
Article in English | MEDLINE | ID: mdl-7805932

ABSTRACT

Retrovirus-like particles were detected by the negative staining method in supernatants of lymph node and spleen cell suspensions prepared from Sprague-Dawley rats with spontaneous acute lymphoblastic leukaemia (ALL). Similar particles were found in supernatants of cell suspensions from a lymphoma that developed after inoculation of lymph node and spleen cell suspension prepared from animals with spontaneous ALL into the subcutis of Sprague-Dawley recipients. On ultrathin sections, budding forms of the virus particles were seen as a dot at the periphery of neoplastically transformed cells.


Subject(s)
Leukemia, Experimental/microbiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/microbiology , Rats, Sprague-Dawley/microbiology , Retroviridae/isolation & purification , Animals , Female , Lymph Nodes/microbiology , Lymphoma/microbiology , Lymphoma/ultrastructure , Male , Microscopy, Electron , Neoplasm Transplantation , Rats , Retroviridae/ultrastructure , Species Specificity , Spleen/microbiology , Tumor Cells, Cultured
12.
J Virol ; 67(12): 7140-8, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8230436

ABSTRACT

Mo+PyF101 M-MuLV is a variant Moloney murine leukemia virus containing polyomavirus F101 enhancers inserted just downstream from the M-MuLV enhancers in the long terminal repeat (LTR). The protein coding sequences for this virus are identical to those of M-MuLV. Mo+PyF101 M-MuLV induces T-cell disease with a much lower incidence and longer latency than wild-type M-MuLV. We have previously shown that Mo+PyF101 M-MuLV is defective in preleukemic events induced by wild-type M-MuLV, including splenic hematopoietic hyperplasia, bone marrow depletion, and generation of recombinant mink cell focus-inducing viruses (MCFs). We also showed that an M-MCF virus driven by the Mo+PyF101 LTR is infectious in vitro but does not propagate in mice. However, in these experiments, when a pseudotypic mixture of Mo+PyF101 M-MuLV and Mo+PyF101 MCF was inoculated into newborn NIH Swiss mice, they died of T-cell leukemia at times almost equivalent to those induced by wild-type M-MuLV. Tumor DNAs from Mo+PyF101 M-MuLV-Mo+PyF101 MCF-inoculated mice were examined by Southern blot analysis. The predominant forms of Mo+PyF101 MCF proviruses in these tumors contained added sequences in the U3 region of the LTR. The U3 regions of representative tumor-derived variant Mo+PyF101 MCFs were cloned by polymerase chain reaction amplification, and sequencing indicated that they had acquired an additional copy of the M-MuLV 75-bp tandem repeat in the enhancer region. NIH 3T3 cell lines infected with altered viruses were obtained from representative Mo+PyF101 M-MuLV-Mo+PyF101 MCF-induced tumors, and mice were inoculated with the recovered viruses. Leukemogenicity was approximately equivalent to that in the original Mo+PyF101 M-MuLV-Mo+PyF101 MCF viral stock. Southern blot analysis on the resulting tumors now predominantly revealed loss of the polyomavirus sequences. These results suggest that the suppressive effects of the PyF101 sequences on M-MuLV-induced disease and potentially on MCF propagation were overcome in two ways: by triplication of the M-MuLV direct repeats and by loss of the polyomavirus sequences.


Subject(s)
Leukemia, Experimental/genetics , Mink Cell Focus-Inducing Viruses/genetics , Moloney murine leukemia virus/genetics , Repetitive Sequences, Nucleic Acid/genetics , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , 3T3 Cells , Animals , Base Sequence , Cloning, Molecular , DNA, Neoplasm/genetics , Genetic Variation , Leukemia, Experimental/etiology , Leukemia, Experimental/microbiology , Leukemia, T-Cell/etiology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/microbiology , Mice , Mice, Inbred Strains , Molecular Sequence Data , Moloney murine leukemia virus/pathogenicity , Proviruses/genetics , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Virulence
13.
J Virol ; 67(10): 6105-9, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8396671

ABSTRACT

Recombinant inbred BXH-2 mice spontaneously produce a B-tropic murine leukemia virus (MuLV) beginning early in life and have a high incidence of spontaneous myeloid leukemia. These traits are not characteristic of the progenitor strains (C57BL/6J and C3H/HeJ) or of 11 other recombinant inbred BXH strains. Genetic analysis has shown that the virus is not transmitted through the germ line, suggesting that the virus is passed from one generation to the next by horizontal transmission. An additional ecotropic proviral locus was detected in some mice of this strain after several generations of inbreeding. We show that BXH ecotropic virus was transmitted to other strains when fostered on viremic BXH-2 mice and that these mice go on to develop tumors of hematopoietic origin. Our earlier finding that virus is expressed early in gestation suggested that the ecotropic MuLV is also transmitted in utero. In order to determine the stage at which the ecotropic MuLV is transmitted in utero, preimplantation stage embryos were transferred to the uteri of recipient ecotropic virus-negative mice. These mice were found to be negative for the presence of the ecotropic MuLV, suggesting that transplacental transmission of the ecotropic virus readily occurs in BXH-2 mice. Although other viruses, including human lentiviruses, are transmitted across the placental barrier, transplacental transmission of MuLV is a rare event. Thus, the BXH-2 mouse strain may contribute to our understanding of the mechanism of transplacental transmission and pathogenesis and offers a potential new model for use in drug therapy of exogenously transmitted viruses related to lentiviruses.


Subject(s)
Leukemia Virus, Murine , Leukemia, Experimental/microbiology , Maternal-Fetal Exchange , Pregnancy Complications, Infectious/microbiology , Animals , Blotting, Southern , Crosses, Genetic , DNA, Viral/genetics , DNA, Viral/isolation & purification , Electrophoresis, Agar Gel , Embryo Transfer , Female , Leukemia Virus, Murine/isolation & purification , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Milk/microbiology , Pregnancy , Recombination, Genetic , Species Specificity
14.
Leuk Res ; 17(9): 741-8, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8371574

ABSTRACT

Based on previous studies where it was shown that non-absorbable antibiotics can influence the normal hematopoiesis via changes in factors related to the intestinal microflora, the influence of vancomycin on the progression of acute myeloid leukemia was investigated in the BNML rat model. Oral vancomycin, which selectively reduces Gram-positive bacteria in the gut, leads to diminution of the leukemic load in liver and spleen by 30-60%. This 'antileukemic effect' is not dependent on Gram-negative bacteria as source for endotoxin. The presumed mechanism is a decrease of the leukemic growth fraction caused by alterations in the absorption of substances from intestinal Gram-positive bacteria.


Subject(s)
Bacteria/drug effects , Intestines/microbiology , Leukemia, Experimental/pathology , Liver/pathology , Spleen/pathology , Vancomycin/pharmacology , Animals , Bromodeoxyuridine/metabolism , Female , Leukemia, Experimental/microbiology , Lipopolysaccharides/analysis , Organ Size/drug effects , Rats , Rats, Inbred BN , Specific Pathogen-Free Organisms
15.
J Virol ; 67(8): 4722-31, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8392610

ABSTRACT

The Graffi murine leukemia virus (MuLV) is a retroviral mixture that induces predominantly myeloid leukemia in several inbred strains of mice. To analyze the viral component responsible for the myeloid leukemogenesis, we cloned several proviruses from a Graffi MuLV-infected cell line. Several infectious molecular clones were obtained that could be classified into two distinct groups of infectious MuLV. Both types of MuLV were nondefective, ecotropic, and NB tropic and induced granulocytic leukemia in BALB/c and NFS mice. Restriction enzyme analysis and molecular hybridization with several MuLV probes on one molecular clone from each group revealed that both groups are closely related to each other but are clearly distinct from all known retroviruses. One component of MuLV, however, induced leukemia with a shorter latency period and harbored a lengthier long terminal repeat. The long terminal repeat of the more leukemogenic component of MuLV had acquired a 60-bp perfect duplication in the U3 region. Analysis of the tumor DNAs with probes for the mouse T-cell receptor and immunoglobulin heavy chain genes revealed frequent rearrangements with one or both probes. This concomitant expression by leukemic cells of markers of different lineages, observed in human leukemias, has been termed "lineage infidelity" and confirms that the latter rearrangements are not restricted to hematopoietic precursors committed to lymphoid differentiation.


Subject(s)
Leukemia Virus, Murine/genetics , Leukemia, Experimental/microbiology , Leukemia, Myeloid/microbiology , Animals , Base Sequence , Blotting, Southern , Cell Line , Cloning, Molecular , DNA Probes , DNA, Viral/genetics , DNA, Viral/isolation & purification , Genes, Immunoglobulin , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/physiopathology , Leukemia, Myeloid/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/genetics , Repetitive Sequences, Nucleic Acid , Restriction Mapping
16.
J Virol ; 67(7): 3763-70, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8510205

ABSTRACT

We analyzed viral recombination events that occur during the preleukemic period in AKR mice. We tagged a molecular chimera between the nonleukemogenic virus Akv and the leukemogenic mink cell focus-inducing (MCF) virus MCF 247 with an amber suppressor tRNA gene, supF. We injected the supF-tagged chimeric virus that contains all of the genes of MCF 247 except the envelope gene, which in turn is derived from Akv, into newborn AKR mice to evaluate its pathogenic potential. Approximately the same percentage of animals developed leukemia with similar latent periods when injected with either the tagged or nontagged virus. DNA from tumors induced in AKR mice by the tagged chimeric virus was analyzed by Southern blotting with the supF gene as a probe. One set of tumors contained the injected supF-tagged virus. Two kinds of supF-tagged proviruses were found in a second set of tumors. One group of supF-tagged viruses had a restriction map consistent with that of the injected virus, while the other group of proviruses had restriction maps that suggested that the proviruses had acquired an MCF virus-like envelope gene by recombination with endogenous viral sequences. These results demonstrate that injected viruses recombine in vivo with endogenous viral sequences. Furthermore, the progression to leukemia was accelerated in mice that develop tumors containing proviruses with an MCF virus env gene, emphasizing the importance of the role of the MCF virus env gene product in transformation.


Subject(s)
AKR murine leukemia virus/genetics , Leukemia, Experimental/microbiology , Mice, Inbred AKR/microbiology , Mink Cell Focus-Inducing Viruses/genetics , Animals , Blotting, Southern , Genes, env , Leukemia, Experimental/genetics , Lymphoma/genetics , Lymphoma/microbiology , Mice , Mink Cell Focus-Inducing Viruses/pathogenicity , Recombination, Genetic , Restriction Mapping , Thymoma/genetics , Thymoma/microbiology
17.
J Leukoc Biol ; 54(1): 40-6, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8336078

ABSTRACT

We examined leukemic cells, HL-60, an acute promyelocytic leukemia cell line, after differentiation induced by 1,25-dihydroxyvitamin D3 (D3) and retinoic acid (A) for infection of Legionella pneumophila, the etiologic agent of Legionnaires' disease. We investigated the effect of interferon gamma (IFN-gamma) on the differentiated cells and on the intracellular growth of the bacteria. An examination of morphological and antigenic changes in the cells was also included in the study. After 4-day incubation with 10(-6)M D3 or A, the HL-60 cells differentiated into monocyte-like (D3-HL-60) or mature granulocyte-like (A-HL-60) cells, respectively. They were then infected with L. pneumophila. Intracellular multiplication of the bacteria was evident in D3-HL-60 cells but not in HL-60 or A-HL-60 cells. D3-HL-60 cells required a 24-h infection time for the intracellular growth of L. pneumophila. D3-HL-60 cells activated with human recombinant IFN-gamma for 1-24 h (gamma-IFN-D3-HL-60 cells) before infection markedly inhibited L. pneumophila multiplication, the effect of IFN-gamma being dose dependent. Surface marker analysis was carried out in HL-60, D3-HL-60, and gamma-IFN-D3-HL-60 cells. On D3-HL-60 cells, CD11b, CD11c, CD14, and CD35 antigen increased, whereas CD71 and HLA-DR antigen decreased. This finding suggested that HL-60 cells differentiated into monocyte-like cells; the acquisition of the complement receptors, CD11b(CR3) and CD35(CR1), seemed to be important for phagocytosis and for the subsequent intracellular multiplication of L. pneumophila. The gamma-IFN-D3-HL-60 cells showed an increase of CD16, CD36, CD71, and HLA-DR antigen, suggesting that they were in an activated state. Our study indicated, first, that D3 can induce human leukemic cells to differentiate into functional monocyte-macrophage-like cells that can support the intracellular multiplication of L. pneumophila and, second, that these differentiated leukemic cells can be activated by IFN-gamma to markedly inhibit bacterial growth.


Subject(s)
Calcitriol/pharmacology , Interferon-gamma/pharmacology , Legionella pneumophila/growth & development , Leukemia, Promyelocytic, Acute/microbiology , Animals , Antigens, Bacterial/analysis , Antigens, CD/analysis , Antigens, Neoplasm/analysis , Cell Differentiation/drug effects , Guinea Pigs , Humans , Intracellular Fluid/microbiology , Legionella pneumophila/drug effects , Legionella pneumophila/immunology , Legionnaires' Disease/drug therapy , Legionnaires' Disease/pathology , Leukemia, Experimental/microbiology , Leukemia, Experimental/pathology , Leukemia, Promyelocytic, Acute/immunology , Leukemia, Promyelocytic, Acute/pathology , Macrophage Activation , Macrophages/cytology , Macrophages/drug effects , Macrophages/physiology , Monocytes/cytology , Monocytes/drug effects , Monocytes/physiology , Recombinant Proteins , Tretinoin/pharmacology , Tumor Cells, Cultured
18.
Leukemia ; 7(7): 1041-6, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8321019

ABSTRACT

Resistance and/or susceptibility for Friend leukemia virus (FLV)-induced leukemogenesis was examined in the fully H-2 incompatible C57BL/6 (B6)-->C3H radiation bone marrow chimeras (RBMC). The results indicated that B6-->C3H chimeras never developed FLV-induced leukemias when infected with FLV 4 months after bone marrow transplantation (BMT). Spleen cells from B6-->C3H chimeras that were preimmunized with 100 Gy-irradiated FBL-3 cells (FLV-induced leukemic cell line originated from B6 mice) were shown to generate anti-FBL-3 specific T-cell proliferation as well as cytotoxic T cells. We also found that when bone marrow cells from B6 mice were mixed with those from C3H mice and then grafted into supralethally irradiated C3H mice, resulting chimeras whose peripheral blood contained less than 30% C3H-derived (susceptible) cells were refractory to FLV-induced leukemogenesis. On the other hand, when C3H mice were infected with FLV and then supralethally irradiated 5 days later and grafted with bone marrow from B6 donors, they developed leukemias which were of B6 origin. Athymic nu/nu mice of B6 background were again shown to develop leukemia following infection with FLV. Possible implication of these findings on the role of T cell-mediated immune response in resistance to FLV-induced leukemogenesis and the immunocompetent nature of fully H-2 incompatible RBMC were discussed.


Subject(s)
Leukemia, Experimental/genetics , Animals , Bone Marrow Transplantation , Friend murine leukemia virus , Leukemia, Experimental/microbiology , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Nude , Radiation Chimera , Survival Analysis , T-Lymphocytes/immunology
19.
Virology ; 195(1): 286-91, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8391185

ABSTRACT

The long terminal repeats (LTRs) of defective Moloney Murine Leukemia Virus (M-MuLV) containing the avian v-myc and v-src oncogenes were exchanged for LTRs from murine retroviruses inducing myeloid and erythroid disease, in an attempt to retarget disease specificity. Chimeric MuLVs containing either the Myeloproliferative sarcoma virus or the Rauscher mink cell focus-inducing virus LTRs induced the same disease as the parental viruses, suggesting that for these viruses the v-myc and v-src oncogenes are the major determinants in the disease specificity. However, substitution of the LTRs did affect the efficiency of tumorigenesis.


Subject(s)
Genes, myc , Genes, src , Leukemia, Experimental/etiology , Moloney murine leukemia virus/physiology , Repetitive Sequences, Nucleic Acid , Animals , Avian Sarcoma Viruses/genetics , Cells, Cultured , Chimera , Cloning, Molecular , DNA, Neoplasm/genetics , Leukemia, Experimental/microbiology , Mice , Moloney murine leukemia virus/genetics , Rauscher Virus/genetics
20.
Oncogene ; 8(6): 1621-30, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8502483

ABSTRACT

The late stages of the erythroleukemias induced by either the replication-defective Friend spleen focus-forming virus (SFFV) or the Friend murine leukemia virus (F-MuLV) are associated with the insertional activation of one of two members (Spi-1 or Fli-1) of the Ets protooncogene family of transcriptional factors. Fli-1 is not rearranged or activated in the erythroleukemias induced by SFFV, and similarly Spi-1 is not rearranged or activated in the leukemic cell clones induced by F-MuLV. This strict specificity of integration sites suggests that Fli-1 and Spi-1 may be functionally distinct and transactivate different downstream genes during the progression of multistage Friend erythroleukemia. In this study, we show that the Fli-1 protein, like other Ets proteins, has DNA-binding activity and can act as a sequence-specific transcriptional activator. We also show that the Fli-1 and Spi-1 proteins are functionally distinct in that they recognize and transactivate through distinct DNA binding sites. Furthermore, we have identified an octanucleotide core sequence that is required in vitro for optimal binding of Fli-1 to the Drosophila E74 target and the promoter sequence of the human GPIIB gene.


Subject(s)
DNA-Binding Proteins/genetics , Friend murine leukemia virus/genetics , Leukemia, Erythroblastic, Acute/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogenes , Sarcoma, Ewing/genetics , Spleen Focus-Forming Viruses/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Base Sequence , Cell Line , DNA-Binding Proteins/metabolism , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/microbiology , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides , Platelet Membrane Glycoproteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Mas , Proto-Oncogene Protein c-fli-1 , Proto-Oncogene Proteins c-ets , Sequence Homology, Nucleic Acid , Trans-Activators/metabolism , Transcription Factors/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...