Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Curr Top Microbiol Immunol ; 313: 3-29, 2007.
Article in English | MEDLINE | ID: mdl-17217036

ABSTRACT

Mouse models of acute promyelocytic leukemia have been generated through transgenic, knock-in, retroviral, and xenograft strategies. These models have been used to elucidate mechanisms underlying leukemogenesis. Among the areas investigated are the role of reciprocal fusions; effects of target cells, expression levels, and mouse strains; cooperating events; and restrictive and permissive factors. These models have also been used to gain insight into the effects of the immune system on leukemic cells and the mechanism of response to retinoic acid. Furthermore, preclinical studies utilizing these mice have advanced therapy for myeloid leukemia.


Subject(s)
Antineoplastic Agents/therapeutic use , Disease Models, Animal , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/physiopathology , Animals , Humans , Mice , Mice, Transgenic
3.
J Virol ; 75(19): 9427-34, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533205

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a disease that occurs in young children and is associated with a high mortality rate. In most patients, JMML has a progressive course leading to death by virtue of infection, bleeding, or progression to acute myeloid leukemia (AML). As it is known that children with neurofibromatosis type 1 syndrome have a markedly increased risk of developing JMML, we have previously developed a mouse model of JMML through reconstitution of lethally irradiated mice with hematopoietic stem cells homozygous for a loss-of-function mutation in the Nf1 gene (D. L. Largaespada, C. I. Brannan, N. A. Jenkins, and N. G. Copeland, Nat. Genet. 12:137-143, 1996). In the course of these experiments, we found that all these genetically identical reconstituted mice developed a JMML-like disorder, but only a subset went on to develop more acute disease. This result strongly suggests that additional genetic lesions are responsible for disease progression to AML. Here, we describe the production of a unique tumor panel, created using the BXH-2 genetic background, for identification of these additional genetic lesions. Using this tumor panel, we have identified a locus, Epi1, which maps 30 to 40 kb downstream of the Myb gene and appears to be the most common site of somatic viral integration in BXH-2 mice. Our findings suggest that proviral integrations at Epi1 cooperate with loss of Nf1 to cause AML.


Subject(s)
Leukemia, Experimental/genetics , Leukemia, Myeloid/genetics , Acute Disease , Animals , Gene Deletion , Genes, Neurofibromatosis 1 , Genes, myb , Genetic Predisposition to Disease , Leukemia, Experimental/etiology , Leukemia, Experimental/virology , Leukemia, Myeloid/etiology , Leukemia, Myeloid/virology , Mice , Retroviridae/genetics , Virus Integration/genetics
4.
Cancer Res ; 61(8): 3472-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309310

ABSTRACT

Mucinous colorectal cancers exhibit a characteristic set of molecular genetic alterations and may be derived from progenitor cells committed to the goblet cell lineage. Previously, we demonstrated that the MUC2 mucin gene promoter drives transgene reporter expression with high specificity in small intestinal goblet cells of transgenic mice. On the basis of these experiments, we reasoned that the MUC2 promoter could be used to drive SV40 T antigen (Tag) expression in the same cell type, decoupling them from their normal antiproliferative controls. A line of mice was established (MUCTag6) that expressed Tag in intestinal goblet cells as determined by RNA blot and immunohistochemical analysis. These goblet cells were markedly involuted however, most notably in the villi. Endogenous intestinal MUC2 message levels were reduced to about one third the normal level in these mice. However, absorptive cell lineage markers were comparable with nontransgenics. Bromodeoxyuridine-positive S-phase cells are limited to crypts in nontransgenic intestine but are present in both crypts and villi in MUCTag6. In contrast, mitotic cells were not present in the villi, indicating that MUCTag6 villi goblet cells do not progress into M phase. Apoptotic cells positive for terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling were increased more than fourfold in MUCTag6 villi (P < 0.0001), and apoptotic goblet cells were evident. Electron microscopic examination of MUCTag6 intestinal villi revealed the presence of degraded cell remnants containing mucin goblets together with other cell debris, further indicating apoptosis of the goblet cell lineage. Thus, the expression of Tag in intestinal goblet cells releases them from normal antiproliferative controls, causing their inappropriate entry into S phase even after they transverse the crypt/villus junction. They do not, however, progress to M phase. Instead, they undergo apoptosis with a high degree of efficiency in S or G(2) phase. These experiments demonstrate that apoptosis effectively blocks inappropriate goblet cell proliferation in the intestine, supporting its proposed role as an antineoplastic mechanism.


Subject(s)
Antigens, Polyomavirus Transforming/biosynthesis , Apoptosis/physiology , Cell Movement/physiology , Goblet Cells/cytology , Intestine, Small/cytology , Mucins/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Female , Goblet Cells/immunology , Goblet Cells/metabolism , Intestine, Small/metabolism , Intestine, Small/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microvilli/physiology , Mucin-2 , Oncogenes , Promoter Regions, Genetic , S Phase/physiology
5.
J Exp Med ; 193(4): 531-43, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11181704

ABSTRACT

The promyelocytic leukemia retinoic acid receptor alpha (PMLRARalpha) chimeric protein is associated with acute promyelocytic leukemia (APL). PMLRARalpha transgenic mice develop leukemia only after several months, suggesting that PMLRARalpha does not by itself confer a fully malignant phenotype. Suppression of apoptosis can have a central role in tumorigenesis; therefore, we assessed whether BCL-2 influenced the ability of PMLRARalpha to initiate leukemia. Evaluation of preleukemic animals showed that whereas PMLRARalpha alone modestly altered neutrophil maturation, the combination of PMLRARalpha and BCL-2 caused a marked accumulation of immature myeloid cells in bone marrow. Leukemias developed more rapidly in mice coexpressing PMLRARalpha and BCL-2 than in mice expressing PMLRARalpha alone, and all mice expressing both transgenes succumbed to leukemia by 7 mo. Although both preleukemic, doubly transgenic mice and leukemic animals had abundant promyelocytes in the bone marrow, only leukemic mice exhibited thrombocytopenia and dissemination of immature cells. Recurrent gain of chromosomes 7, 8, 10, and 15 and recurrent loss of chromosome 2 were identified in the leukemias. These chromosomal changes may be responsible for the suppression of normal hematopoiesis and dissemination characteristic of the acute leukemias. Our results indicate that genetic changes that inhibit apoptosis can cooperate with PMLRARalpha to initiate APL.


Subject(s)
Leukemia, Promyelocytic, Acute/etiology , Neoplasm Proteins/metabolism , Neutrophils/cytology , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Antigens, Differentiation/genetics , Apoptosis/genetics , Bone Marrow Cells/cytology , Calcium-Binding Proteins/genetics , Calgranulin A , Cell Differentiation , Cell Division , Cell Transformation, Neoplastic , Chromosome Aberrations , Chromosome Disorders , Hematopoietic Stem Cells , Leukemia, Promyelocytic, Acute/mortality , Leukemia, Promyelocytic, Acute/pathology , Leukopoiesis , Mice , Mice, Transgenic , Myeloid Cells/cytology , Recombinant Fusion Proteins/metabolism
6.
Blood ; 95(5): 1541-50, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10688806

ABSTRACT

The most common chromosomal translocation in acute promyelocytic leukemia (APL), t15;17(q22;q21), creates PMLRARalpha and RARalphaPML fusion genes. We previously developed a mouse model of APL by expressing PMLRARalpha in murine myeloid cells. In order to examine the mechanisms by which PMLRARalpha can initiate leukemia, we have now generated transgenic mice expressing PMLRARalpham4 and RARalpham4, proteins that are unable to activate transcription in response to retinoic acid. PMLRARalpham4 transgenic mice developed myeloid leukemia, demonstrating that transcriptional activation by PMLRARalpha is not required for leukemic transformation. The characteristics of the leukemias arising in the PMLRARalpham4 transgenic mice varied from those previously observed in our PMLRARalpha transgenic mice, indicating that ligand responsiveness may influence the phenotype of the leukemic cells. The leukemias that arose in PMLRARalpham4 transgenic mice did not differentiate in response to retinoic acid therapy. This result supports the hypothesis that a major therapeutic effect of retinoic acid is mediated directly through the PMLRARalpha protein. However, a variable effect on survival suggested that this agent may be of some benefit in APL even when leukemic cells are resistant to its differentiative effects. Transgenic mice expressing high levels of RARalpham4 have not developed leukemia, providing evidence that the PML domain of PMLRARalpha plays a specific and critical role in the pathogenesis of APL. (Blood. 2000;95:1541-1550)


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Experimental/genetics , Leukemia, Promyelocytic, Acute/metabolism , Neoplasm Proteins/chemistry , Oncogene Proteins, Fusion/chemistry , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Chlorocebus aethiops , Disease Progression , Genes, Dominant , Humans , Leukemia, Experimental/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Transgenic , Mutagenesis , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , Phenotype , Protein Structure, Tertiary , Radiation Chimera , Recombinant Fusion Proteins/physiology , Repressor Proteins/physiology , Transfection , Tretinoin/therapeutic use
7.
Blood Cells Mol Dis ; 26(6): 620-5, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11358354

ABSTRACT

Murine models of human neoplasms are being used to expand our understanding of pathogenesis and to develop improved cancer therapies. MRP8-PMLRARalpha transgenic mice represent one model of human acute promyelocytic leukemia (APL). These mice develop leukemias that mirror characteristic features of human APL including responsiveness to retinoic acid and arsenic. This model is proving its value in elucidating mechanisms by which PMLRARalpha contributes to leukemia, identifying genetic changes that cooperate to cause leukemia, and investigating new molecular targets for leukemia therapy. These studies suggest that acute myeloid leukemias (AMLs) such as APL result from genetic changes that combine to both impair differentiation and allow immature cells to survive and proliferate outside of a normal microenvironment. Retinoic acid targets the central molecular lesion in human APL and has greatly improved survival. Molecularly targeted therapies that either restore maturation or abrogate growth autonomy represent a hope for improving survival of patients with other subtypes of AML.


Subject(s)
Disease Models, Animal , Leukemia, Promyelocytic, Acute , Animals , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Leukemia, Promyelocytic, Acute/therapy , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics
10.
J Exp Med ; 189(7): 1043-52, 1999 Apr 05.
Article in English | MEDLINE | ID: mdl-10190895

ABSTRACT

In acute promyelocytic leukemia (APL) patients, retinoic acid (RA) triggers differentiation while arsenic trioxide (arsenic) induces both a partial differentiation and apoptosis. Although their mechanisms of action are believed to be distinct, these two drugs both induce the catabolism of the oncogenic promyelocytic leukemia (PML)/RARalpha fusion protein. While APL cell lines resistant to one agent are sensitive to the other, the benefit of combining RA and arsenic in cell culture is controversial, and thus far, no data are available in patients. Using syngenic grafts of leukemic blasts from PML/RARalpha transgenic mice as a model for APL, we demonstrate that arsenic induces apoptosis and modest differentiation, and prolongs mouse survival. Furthermore, combining arsenic with RA accelerates tumor regression through enhanced differentiation and apoptosis. Although RA or arsenic alone only prolongs survival two- to threefold, associating the two drugs leads to tumor clearance after a 9-mo relapse-free period. These studies establishing RA/arsenic synergy in vivo prompt the use of combined arsenic/RA treatments in APL patients and exemplify how mouse models of human leukemia can be used to design or optimize therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Arsenic/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Tretinoin/pharmacology , Animals , Apoptosis/drug effects , Arsenic/administration & dosage , Cell Differentiation/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Hematopoiesis/drug effects , Humans , Leukemia, Promyelocytic, Acute/pathology , Liver/pathology , Lung/pathology , Mice , Mice, Transgenic , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Remission Induction , Spleen/pathology , Tretinoin/administration & dosage , Tumor Cells, Cultured
11.
Proc Natl Acad Sci U S A ; 95(20): 11863-8, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751756

ABSTRACT

Chromosomal translocations involving the genes encoding the alpha and beta subunits of the Pebp2/Cbf transcription factor have been associated with human acute myeloid leukemia and the preleukemic condition, myelodysplasia. Inv(16)(p13;q22) fuses the gene encoding the beta subunit of Pebp2 to the MYH11 gene encoding a smooth muscle myosin heavy chain (Smmhc). To examine the effect of the inv(16)(p13;q22) on myelopoiesis, we used the hMRP8 promoter element to generate transgenic mice expressing the Pebp2betaSmmhc chimeric fusion protein in myeloid cells. Neutrophil maturation was impaired in PEBP2betaMYH11 transgenic mice. Although the transgenic mice had normal numbers of circulating neutrophils, their bone marrow contained increased numbers of immature neutrophilic cells, which exhibited abnormal characteristics. In addition, PEBP2betaMYH11 inhibited neutrophilic differentiation in colonies derived from hematopoietic progenitors. Coexpression of both PEBP2betaMYH11 and activated NRAS induced a more severe phenotype characterized by abnormal nuclear morphology indicative of granulocytic dysplasia. These results show that PEBP2betaMYH11 can impair neutrophil development and provide evidence that alterations of Pebp2 can contribute to the genesis of myelodysplasia.


Subject(s)
Chromosome Inversion , Chromosomes, Human, Pair 16/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Neutrophils/pathology , Oncogene Proteins, Fusion/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , DNA-Binding Proteins/physiology , Gene Expression , Granulocytes/pathology , Hematopoiesis/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Leukocyte Count , Mice , Mice, Transgenic , Myelodysplastic Syndromes/etiology , Myelodysplastic Syndromes/genetics , Oncogene Proteins, Fusion/physiology , Transcription Factor AP-2 , Transcription Factors/physiology
12.
EMBO J ; 14(3): 461-72, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7859736

ABSTRACT

Tissue homeostasis and the prevention of neoplasia require regulatory co-ordination between cellular proliferation and apoptosis. Several cellular proteins, including c-myc and E2F, as well as viral proteins such as E1A, have dual functions as positive regulators of apoptosis and proliferation. The product of the retinoblastoma tumor suppressor gene, pRb, binds these proteins and is known to function in growth suppression. To examine whether pRb may function as a negative regulator of both proliferation and apoptosis, we analyzed apoptosis induced in transfected derivatives of the human osteosarcoma cell line SAOS-2. Ionizing radiation induced apoptosis in a time- and dose-dependent manner in SAOS-2 cells, which lack pRb expression. In both a transient and stable transfection assay, SAOS-2 derivatives expressing wild-type (wt) pRb exhibited increased viability and decreased apoptosis following treatment at a variety of radiation doses. Expression in SAOS-2 of a mutant pRb that fails to complex with several known binding partners of pRb, including E1A and E2F, did not protect SAOS-2 cells from apoptosis. Radiation exposure induced a G2 arrest in SAOS-2 and in derivatives expressing pRb. Inhibition of DNA synthesis and cell cycle progression by aphidicolin treatment failed to protect SAOS-2 cells or pRb-expressing isolates from undergoing apoptosis. Our data document a novel function for pRb in suppressing apoptosis and suggest that several proteins shown to induce apoptosis, including E1A, E2F and c-myc, may do so by interfering with the protective function of pRb.


Subject(s)
Apoptosis/physiology , Genes, Retinoblastoma/physiology , Radiation Tolerance/physiology , Retinoblastoma Protein/physiology , Aphidicolin/pharmacology , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , Dose-Response Relationship, Radiation , Genes, Retinoblastoma/genetics , Humans , Osteosarcoma , Radiation Tolerance/genetics , Retinoblastoma Protein/genetics , Transfection , Tumor Cells, Cultured , X-Rays/adverse effects
13.
N Engl J Med ; 317(16): 985-90, 1987 Oct 15.
Article in English | MEDLINE | ID: mdl-3657865

ABSTRACT

We report the development of a rapid nonradioactive technique for the genetic prediction of human disease and its diagnostic application to hemophilia A. This method is based on enzymatic amplification of short segments of human genes associated with inherited disorders. A novel feature of the procedure is the use of a heat-stable DNA polymerase, which allows the repeated rounds of DNA synthesis to proceed at 63 degrees C. The high sequence specificity of the amplification reaction at this elevated temperature permits restriction-site polymorphisms, contained in the amplified samples, to be analyzed by visual inspection of their digestion products on polyacrylamide gels. By means of this method, we have performed carrier detection and prenatal diagnosis of hemophilia in two families with use of the factor VIII intragenic polymorphisms identified by the restriction enzymes BclI and XbaI. Predictions can be made directly from chorionic villi, without previous DNA extraction, and fetal sex can be determined by amplification of sequences specific for the Y chromosome. Specific amplification of genomic sequences with heat-stable DNA polymerase is applicable to the diagnosis of a wide variety of inherited disorders. These include diseases diagnosed by restriction-site variation, such as Duchenne's muscular dystrophy and sickle cell anemia, those due to a collection of known mutations, such as beta-thalassemia, and those due to gene deletion, such as alpha-thalassemia.


Subject(s)
DNA/analysis , Fetal Diseases/diagnosis , Genetic Diseases, Inborn/diagnosis , Hemophilia A/diagnosis , Nucleic Acid Amplification Techniques , Prenatal Diagnosis/methods , Base Sequence , DNA-Directed DNA Polymerase/pharmacology , Female , Genetic Carrier Screening/methods , Humans , Male , Pedigree , Polymorphism, Genetic , Pregnancy , Sex Determination Analysis , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...