Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Leuk Res ; 38(3): 377-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24461365

ABSTRACT

The nitric oxide (NO) prodrug JS-K, a promising anti-cancer agent, consists of a diazeniumdiolate group necessary for the release of NO as well as an arylating ring. In this study, we research the mechanism by which JS-K kills a murine erythroleukemia cell line and determine the roles of NO and arylation in the process. Our studies indicate that JS-K inhibits the PI 3-kinase/Akt and MAP kinase pathways. This correlates with the activation of the tumor suppressor FoxO3a and increased expression of various caspases, leading to apoptosis. The arylating capability of JS-K appears to be sufficient for inducing these biological effects. Overall, these data suggest that JS-K kills tumor cells by arylating and inactivating signaling molecules that block the activation of a tumor suppressor.


Subject(s)
Azo Compounds/pharmacology , Cytotoxins/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Nitric Oxide Donors/pharmacology , Piperazines/pharmacology , Prodrugs/pharmacology , Animals , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Forkhead Box Protein O3 , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
2.
Leuk Res ; 36(3): 369-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21924771

ABSTRACT

Lack of suitable mouse models for central nervous system (CNS)-associated leukemias has hindered mechanism-guided development of therapeutics. By transplanting retrovirus-transformed mouse erythroleukemia cells into syngeneic mice, we developed a new animal model of meningeal leukemia associated with rapid paralysis. Necropsy revealed massive proliferation of the leukemic cells in the bone marrow (BM) followed by pathological angiogenesis and invasion of the leukemic cells into the meninges of the CNS. Further analysis demonstrated that the erythroleukemia cells secreted high levels of VEGF and preferentially adhered in vitro to fibronectin. This unique animal model for meningeal leukemia should facilitate studies of engraftment and proliferation of leukemic cells in the BM and their invasion of the CNS as well as pre-clinical evaluation of experimental therapeutics for CNS-associated leukemias.


Subject(s)
Biomarkers, Tumor/metabolism , Central Nervous System Neoplasms/pathology , Disease Models, Animal , Leukemia, Erythroblastic, Acute/physiopathology , Leukemia, Experimental/pathology , Meningeal Neoplasms/pathology , Retroviridae/genetics , Animals , Biomarkers, Tumor/genetics , Blotting, Western , Cell Adhesion , Cell Proliferation , Central Nervous System Neoplasms/blood supply , Central Nervous System Neoplasms/etiology , Enzyme-Linked Immunosorbent Assay , Fibronectins/metabolism , Gene Expression Profiling , Integrin alpha5beta1/metabolism , Leukemia, Experimental/etiology , Meningeal Neoplasms/blood supply , Meningeal Neoplasms/etiology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Vascular Endothelial Growth Factor A/metabolism
3.
Int J Cancer ; 131(5): 1083-94, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22034044

ABSTRACT

Infection of erythroid cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia in mice, due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator, erythropoietin, because of the interaction among the viral envelope protein, the erythropoietin receptor, and a short form of the receptor tyrosine kinase Stk (sf-Stk). This leads to constitutive activation of several signal transduction pathways. Our previous studies showed that sf-Stk interacts with SFFV gp55, forming disulfide-linked complexes. This covalent interaction, along with other noncovalent interactions with SFFV-gp55, results in constitutive tyrosine phosphorylation of sf-Stk and rodent fibroblast transformation. Here, we determined the precise amino acid region within sf-Stk that contributes to fibroblast transformation by the polycythemia-inducing (SFFV-P) and the anemia-inducing (SFFV-A) strains of SFFV. Sf-Stk deletion mutants showed different transforming abilities in fibroblasts infected with SFFV-P and SFFV-A, although the N-terminal extracellular domain of sf-Stk was essential for fibroblast transformation by both viruses. Point mutations of sf-Stk indicated that cysteine 19 was critical for fibroblast transformation by SFFV-P, although all four cysteines (8, 19, 37 and 42) appeared to be important for fibroblast transformation by both SFFV-P and SFFV-A. Mutation of sf-Stk cysteine 19 abolished its ability to form dimers with SFFV-P and SFFV-A gp55. These results suggest that the interaction between sf-Stk and the envelope proteins of the polycythemia- and anemia-inducing variants of SFFV is architecturally different.


Subject(s)
Anemia/etiology , Cell Transformation, Neoplastic/pathology , Fibroblasts/pathology , Leukemia, Experimental/genetics , Polycythemia/etiology , Receptor Protein-Tyrosine Kinases/metabolism , Retroviridae Infections/genetics , Spleen Focus-Forming Viruses/genetics , Tumor Virus Infections/genetics , Amino Acid Sequence , Anemia/metabolism , Anemia/pathology , Animals , Blotting, Western , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/virology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Phosphorylation , Plasmids/genetics , Polycythemia/metabolism , Polycythemia/pathology , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Sequence Homology, Amino Acid , Signal Transduction , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
5.
Viruses ; 3(12): 2442-61, 2011 12.
Article in English | MEDLINE | ID: mdl-22355448

ABSTRACT

The cell lines of the NCI-60 panel represent different cancer types and have been widely utilized for drug screening and molecular target identification. Screening these cell lines for envelope proteins or gene sequences related to xenotropic murine leukemia viruses (X-MLVs) revealed that one cell line, EKVX, was a candidate for production of an infectious gammaretrovirus. The presence of a retrovirus infectious to human cells was confirmed by the cell-free transmission of infection to the human prostate cancer cell line LNCaP. Amplification and sequencing of additional proviral sequences from EKVX confirmed a high degree of similarity to X-MLV. The cell line EKVX was established following passage of the original tumor cells through nude mice, providing a possible source of the X-MLV found in the EKVX cells.


Subject(s)
Adenocarcinoma/virology , Leukemia Virus, Murine/metabolism , Lung Neoplasms/virology , Animals , Base Sequence , Cell Line, Tumor , Genes, Viral/genetics , Humans , Immunoblotting , Leukemia Virus, Murine/genetics , Leukemia, Experimental/virology , Mice , Mice, Nude/virology , Molecular Sequence Data , Polymerase Chain Reaction , Retroviridae Infections/virology , Sequence Alignment , Tumor Virus Infections/virology , Viral Envelope Proteins/genetics
6.
Virology ; 409(2): 198-203, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21051067

ABSTRACT

We recently reported that infection of rats with the neurodegenerative disease-causing retrovirus PVC-211 MuLV results in elevated levels of the chemokine MIP-1α followed by the accumulation of activated microglia in the brain. To investigate the importance of MIP-1α in recruitment of microglia to the brain, we treated rats with MIP-1α antibodies before and after PVC-211 MuLV infection. This caused a delay in the development of paralysis which was associated with a decrease in activated microglia without affecting virus expression. To determine the source of activated microglia, rats were splenectomized 4 days after virus infection. Splenectomized rats showed a delay in disease development that was associated with decreased numbers of activated microglia without affecting virus expression. Together, these results suggest that MIP-1α is directly involved in the neurodegeneration induced in rats by PVC-211 MuLV by recruiting macrophages/microglia from the periphery into regions of the brain that eventually become diseased.


Subject(s)
Leukemia Virus, Murine/pathogenicity , Macrophage Inflammatory Proteins/immunology , Macrophages/immunology , Neurodegenerative Diseases/pathology , Retroviridae Infections/pathology , Animals , Leukemia Virus, Murine/immunology , Microglia/immunology , Microglia/pathology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/virology , Paralysis/pathology , Paralysis/virology , Rats , Retroviridae Infections/immunology , Retroviridae Infections/virology
7.
J Virol ; 84(15): 7675-82, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20504929

ABSTRACT

Infection of erythroid cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia in mice due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator, erythropoietin (Epo), because of interaction of the viral envelope protein with the erythropoietin receptor and a short form of the receptor tyrosine kinase Stk (sf-Stk), leading to constitutive activation of several signal transduction pathways. Our previous in vitro studies showed that phosphatidylinositol 3-kinase (PI3-kinase) is activated in SFFV-infected cells and is important in mediating the biological effects of the virus. To determine the role of PI3-kinase in SFFV-induced disease, mice deficient in the p85alpha regulatory subunit of class IA PI3-kinase were inoculated with different strains of SFFV. We observed that p85alpha status determined the extent of erythroid hyperplasia induced by the sf-Stk-dependent viruses SFFV-P (polycythemia-inducing strain of SFFV) and SFFV-A (anemia-inducing strain of SFFV) but not by the sf-Stk-independent SFFV variant BB6. Our data also indicate that p85alpha status determines the response of mice to stress erythropoiesis, consistent with a previous report showing that SFFV uses a stress erythropoiesis pathway to induce erythroleukemia. We further showed that sf-Stk interacts with p85alpha and that this interaction depends upon sf-Stk kinase activity and tyrosine 436 in the multifunctional docking site. Pharmacological inhibition of PI3-kinase blocked proliferation of primary erythroleukemia cells from SFFV-infected mice and the erythroleukemia cell lines derived from them. These results indicate that p85alpha may regulate sf-Stk-dependent erythroid proliferation induced by SFFV as well as stress-induced erythroid hyperplasia.


Subject(s)
Leukemia, Erythroblastic, Acute/virology , Phosphatidylinositol 3-Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Spleen Focus-Forming Viruses/pathogenicity , Animals , Cell Line, Tumor , Mice , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinases/deficiency
8.
Viruses ; 2(10): 2235-2257, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21994618

ABSTRACT

HEMATOLOGICAL MALIGNANCIES IN HUMANS TYPICALLY INVOLVE TWO TYPES OF GENETIC CHANGES: those that promote hematopoietic cell proliferation and survival (often the result of activation of tyrosine kinases) and those that impair hematopoietic cell differentiation (often the result of changes in transcription factors). The multi-stage erythroleukemia induced in mice by Friend spleen focus-forming virus (SFFV) is an excellent animal model for studying the molecular basis for both of these changes. Significant progress has been made in understanding the molecular basis for the multi-stage erythroleukemia induced by Friend SFFV. In the first stage of leukemia, the envelope protein encoded by SFFV interacts with and activates the erythropoietin (Epo) receptor and the receptor tyrosine kinase sf-Stk in erythroid cells, causing their Epo-independent proliferation, differentiation and survival. In the second stage, SFFV integration into the Sfpi1 locus activates the myeloid transcription factor PU.1, blocking erythroid cell differentiation, and in conjunction with the loss of p53 tumor suppressor activity, results in the outgrowth of malignant cells. In this review, we discuss the current level of understanding of how SFFV alters the growth and differentiation of erythroid cells and results in the development of erythroleukemia. Our knowledge of how SFFV causes erythroleukemia in mice may give us clues as to how the highly related human retrovirus XMRV causes malignancies in humans.

9.
Science ; 326(5952): 585-9, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19815723

ABSTRACT

Chronic fatigue syndrome (CFS) is a debilitating disease of unknown etiology that is estimated to affect 17 million people worldwide. Studying peripheral blood mononuclear cells (PBMCs) from CFS patients, we identified DNA from a human gammaretrovirus, xenotropic murine leukemia virus-related virus (XMRV), in 68 of 101 patients (67%) as compared to 8 of 218 (3.7%) healthy controls. Cell culture experiments revealed that patient-derived XMRV is infectious and that both cell-associated and cell-free transmission of the virus are possible. Secondary viral infections were established in uninfected primary lymphocytes and indicator cell lines after their exposure to activated PBMCs, B cells, T cells, or plasma derived from CFS patients. These findings raise the possibility that XMRV may be a contributing factor in the pathogenesis of CFS.


Subject(s)
Fatigue Syndrome, Chronic/virology , Gammaretrovirus/isolation & purification , Leukocytes, Mononuclear/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Antibodies, Viral/blood , B-Lymphocytes/immunology , B-Lymphocytes/virology , Base Sequence , Cell Line , Cell Line, Tumor , Coculture Techniques , DNA/genetics , Gammaretrovirus/genetics , Gammaretrovirus/immunology , Gammaretrovirus/physiology , Gene Products, env/analysis , Gene Products, gag/analysis , Genome, Viral , Humans , Lymphocyte Activation , Male , Mice , Molecular Sequence Data , Prostatic Neoplasms/virology , Retroviridae Infections/epidemiology , Retroviridae Infections/transmission , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tumor Virus Infections/epidemiology , Tumor Virus Infections/transmission
10.
J Virol ; 83(10): 4912-22, 2009 May.
Article in English | MEDLINE | ID: mdl-19279110

ABSTRACT

PVC-211 murine leukemia virus (MuLV) is a neuropathogenic retrovirus that has undergone genetic changes from its nonneuropathogenic parent, Friend MuLV, that allow it to efficiently infect rat brain capillary endothelial cells (BCEC). To clarify the mechanism by which PVC-211 MuLV expression in BCEC induces neurological disease, we examined virus-infected rats at various times during neurological disease progression for vascular and inflammatory changes. As early as 2 weeks after virus infection and before any marked appearance of spongiform neurodegeneration, we detected vessel leakage and an increase in size and number of vessels in the areas of the brain that eventually become diseased. Consistent with these findings, the amount of vascular endothelial growth factor (VEGF) increased in the brain as early as 1 to 2 weeks postinfection. Also detected at this early disease stage was an increased level of macrophage inflammatory protein 1 alpha (MIP-1 alpha), a cytokine involved in recruitment of microglia to the brain. This was followed at 3 weeks postinfection by a marked accumulation of activated microglia in the spongiform areas of the brain accompanied by an increase in tissue plasminogen activator, a product of microglia implicated in neurodegeneration. Pathological observations at the end stage of the disease included loss of neurons, decreased myelination, and mild muscle atrophy. Treatment of PVC-211 MuLV-infected rats with clodronate-containing liposomes, which specifically kill microglia, significantly blocked neurodegeneration. Together, these results suggest that PVC-211 MuLV infection of BCEC results in the production of VEGF and MIP-1 alpha, leading to the vascular changes and microglial activation necessary to cause neurodegeneration.


Subject(s)
Chemokine CCL3/metabolism , Leukemia Virus, Murine/pathogenicity , Microglia/virology , Nerve Degeneration/virology , Retroviridae Infections/virology , Vascular Endothelial Growth Factor A/metabolism , Animals , Capillaries/virology , Cells, Cultured , Cerebellum/blood supply , Cerebellum/pathology , Cerebellum/virology , Clodronic Acid/pharmacology , Demyelinating Diseases/virology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Inflammation/virology , Leukemia, Experimental/virology , Microglia/metabolism , Muscular Atrophy/virology , Nerve Degeneration/pathology , Nerve Degeneration/prevention & control , Rats , Rats, Inbred F344 , Tumor Virus Infections/virology
11.
Epigenetics ; 3(3): 134-42, 2008.
Article in English | MEDLINE | ID: mdl-18487951

ABSTRACT

Hematopoietic malignancies are frequently associated with DNA hypomethylation but the molecular mechanisms involved in tumor formation remain poorly understood. Here we report that mice lacking Lsh develop leukemia associated with DNA hypomethylation and oncogene activation. Lsh is a member of the SNF2 chromatin remodeling family and is required for de novo methylation of genomic DNA. Mice that received Lsh deficient hematopoietic progenitors showed severe impairment of hematopoiesis, suggesting that Lsh is necessary for normal hematopoiesis. A subset of mice developed erythroleukemia, a tumor that does not spontaneously occur in mice. Tumor tissues were CpG hypomethylated and showed a modest elevation of the transcription factor PU.1, an oncogene that is crucial for Friend virus induced erythroleukemia. Analysis of Lsh(-/-) hematopoietic progenitors revealed widespread DNA hypomethylation at repetitive sequences and hypomethylation at specific retroviral elements within the PU.1 gene. Wild type cells showed Lsh and Dnmt3b binding at the retroviral elements located within the PU.1 gene. On the other hand, Lsh deficient cells had no detectable Dnmt3b association suggesting that Lsh is necessary for recruitment of Dnmt3b to its target. Furthermore, Lsh(-/-) hematopoietic precursors showed impaired suppression of retroviral elements in the PU.1 gene, an increase of PU.1 transcripts and protein levels. Thus DNA hypomethylation caused by Lsh depletion is linked to transcriptional upregulation of retroviral elements and oncogenes such as PU.1 which in turn may promote the development of erythroleukemia in mice.


Subject(s)
DNA Helicases/deficiency , DNA Methylation , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Animals , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Helicases/genetics , Gene Expression Regulation , Hematopoiesis/genetics , Mice , Mice, Inbred C57BL , Protein Binding , Proto-Oncogene Proteins/genetics , Retroviridae , Trans-Activators/genetics , DNA Methyltransferase 3B
12.
J Virol ; 82(1): 419-27, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17959667

ABSTRACT

Infection of erythroid progenitor cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia and eventually to erythroleukemia in susceptible strains of mice. The viral envelope protein, SFFV gp55, forms a complex with the erythropoietin receptor (EpoR) and a short form of the receptor tyrosine kinase Stk (sf-Stk), activating both and inducing Epo-independent proliferation. Recently, we discovered that coexpression of SFFV gp55 and sf-Stk is sufficient to transform NIH 3T3 and primary fibroblasts. In the current study, we demonstrate that sf-Stk and its downstream effectors are critical to this transformation. Unlike SFFV-derived erythroleukemia cells, which depend on PU.1 expression for maintenance of the transformed phenotype, SFFV gp55-sf-Stk-transformed fibroblasts are negative for PU.1. Underscoring the importance of sf-Stk to fibroblast transformation, knockdown of sf-Stk abolished the ability of these cells to form anchorage-independent colonies. Like SFFV-infected erythroid cells, SFFV gp55-sf-Stk-transformed fibroblasts express high levels of phosphorylated MEK, ERK, phosphatidylinositol 3-kinase (PI3K), Gab1/2, Akt, Jun kinase (JNK), and STAT3, but unlike virus-infected erythroid cells they fail to express phosphorylated STATs 1 and 5, which may require involvement of the EpoR. In addition, the p38 mitogen-activated protein kinase (MAPK) stress response is suppressed in the transformed fibroblasts. Inhibition of either JNK or the PI3K pathway decreases both monolayer proliferation and anchorage-independent growth of the transformed fibroblasts as does the putative kinase inhibitor luteolin, but inhibition of p38 MAPK has no effect. Our results indicate that sf-Stk is a molecular endpoint of transformation that could be targeted directly or with agents against its downstream effectors.


Subject(s)
Cell Transformation, Viral/physiology , Fibroblasts/virology , Receptor Protein-Tyrosine Kinases/physiology , Spleen Focus-Forming Viruses/physiology , Viral Envelope Proteins/physiology , Animals , Cell Proliferation , Gene Silencing , Mice , NIH 3T3 Cells , Protein Kinases/biosynthesis
13.
J Virol ; 80(12): 5678-85, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731906

ABSTRACT

Infection of mice with Friend spleen focus-forming virus (SFFV) results in a multistage erythroleukemia. In the first stage, the SFFV envelope glycoprotein interacts with the erythropoietin receptor and a short form of the receptor tyrosine kinase sf-Stk, resulting in constitutive activation of signal transducing molecules and the development of erythropoietin (Epo)-independent erythroid hyperplasia and polycythemia. The second stage results from the outgrowth of a rare virus-infected erythroid cell that expresses nonphysiological levels of the myeloid transcription factor PU.1. These cells exhibit a differentiation block and can be grown as murine erythroleukemia (MEL) cell lines. In this study, we examined SFFV MEL cells to determine whether their transformed phenotype was associated with a block in the activation of any Epo signal-transducing molecules. Our studies indicate that Epo- or SFFV-induced activation of STAT1/3 DNA binding activity is blocked in SFFV MEL cells. The block is at the level of tyrosine phosphorylation of STAT1, although Jak2 phosphorylation is not blocked in these cells. In contrast to Epo, alpha interferon can induce STAT1 phosphorylation and DNA binding in SFFV MEL cells. The SFFV-transformed cells were shown to express elevated levels of the hematopoietic phosphatase SHP-1, and treatment of the cells with a phosphatase inhibitor restored STAT1 tyrosine phosphorylation. MEL cells derived from Friend murine leukemia virus (MuLV) or ME26 MuLV-infected mice, which do not express PU.1, express lower levels of SHP-1 and are not blocked in STAT1/3 DNA-binding activity. Our studies suggest that SFFV-infected erythroid cells become transformed when differentiation signals activated by STAT1/3 are blocked due to high SHP-1 levels induced by inappropriate expression of the PU.1 protein.


Subject(s)
Cell Transformation, Viral , DNA/metabolism , Erythroblasts/virology , Intracellular Signaling Peptides and Proteins/genetics , Protein Tyrosine Phosphatases/genetics , STAT1 Transcription Factor/metabolism , Spleen Focus-Forming Viruses/pathogenicity , Animals , Cell Differentiation , Erythropoietin/physiology , Gene Expression Regulation , Mice , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
14.
Proc Natl Acad Sci U S A ; 102(43): 15488-93, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16223879

ABSTRACT

Friend spleen focus-forming virus (SFFV) causes rapid erythroleukemia in mice due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator erythropoietin (Epo) because of constitutive activation of Epo signal transduction pathways. Although SFFV infects many cell types, deregulation of cell growth occurs only when SFFV infects erythroid cells, suggesting that these cells express unique proteins that the virus requires to mediate its biological effects. Not only do erythroid cells express the Epo receptor (EpoR), but those from mice susceptible to SFFV-induced erythroleukemia also express a short form of the receptor tyrosine kinase Stk (sf-Stk). In erythroid cells, SFFV gp55 interacts with the EpoR complex and sf-Stk, leading to activation of the kinase and constitutive activation of signal transducing molecules. In this study, we demonstrate that SFFV gp55 can also deregulate the growth of nonerythroid cells when it is coexpressed with sf-Stk. Expression of SFFV gp55 in rodent fibroblasts engineered to express sf-Stk induced their transformation, as demonstrated by focus formation and anchorage-independent growth in vitro. This transformation by SFFV gp55 requires the kinase activity of sf-Stk and the presence of its extracellular domain but not expression of the EpoR or the tyrosine kinase Jak2, which is required for activation of signal transduction pathways through the EpoR. Thus, expression of SFFV gp55 in nonerythroid cells coexpressing sf-Stk results in their uncontrolled growth, demonstrating a previously unrecognized mechanism for retrovirus transformation of rodent fibroblasts and providing insight into SFFV-induced disease.


Subject(s)
Cell Transformation, Viral , Receptor Protein-Tyrosine Kinases/physiology , Spleen Focus-Forming Viruses/pathogenicity , Animals , Fibroblasts/cytology , Janus Kinase 2 , Mice , NIH 3T3 Cells , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/chemistry , Viral Envelope Proteins/physiology
15.
J Virol ; 79(20): 12752-62, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16188978

ABSTRACT

Members of the mitogen-activated protein kinase (MAPK) family, including Jun amino-terminal kinase (JNK) and extracellular signal-related kinase (ERK), play an important role in the proliferation of erythroid cells in response to erythropoietin (Epo). Erythroid cells infected with the Friend spleen focus-forming virus (SFFV) proliferate in the absence of Epo and show constitutive activation of Epo signal transduction pathways. We previously demonstrated that the ERK pathway was constitutively activated in Friend SFFV-infected erythroid cells, and in this study JNK is also shown to be constitutively activated. Pharmacological inhibitors of both the ERK and JNK pathways stopped the proliferation of primary erythroleukemic cells from Friend SFFV-infected mice, with little induction of apoptosis, and furthermore blocked their ability to form Epo-independent colonies. However, only the JNK inhibitor blocked the proliferation of erythroleukemia cell lines derived from these mice. The JNK inhibitor caused significant apoptosis in these cell lines as well as an increase in the fraction of cells in G(2)/M and undergoing endoreduplication. In contrast, the growth of erythroleukemia cell lines derived from Friend murine leukemia virus (MuLV)-infected mice was inhibited by both the MEK and JNK inhibitors. JNK is important for AP1 activity, and we found that JNK inhibitor treatment reduced AP1 DNA-binding activity in primary erythroleukemic splenocytes from Friend SFFV-infected mice and in erythroleukemia cell lines from Friend MuLV-infected mice but did not alter AP1 DNA binding in erythroleukemia cell lines from Friend SFFV-infected mice. These data suggest that JNK plays an important role in cell proliferation and/or the survival of erythroleukemia cells.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Experimental/metabolism , Retroviridae Infections/metabolism , Signal Transduction , Spleen Focus-Forming Viruses/physiology , Tumor Virus Infections/metabolism , Animals , Anthracenes/pharmacology , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/pharmacology , Cell Line, Transformed/virology , Cell Proliferation , Cell Transformation, Viral , Cells, Cultured , Erythropoietin , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured/physiology
16.
J Virol ; 78(9): 4573-81, 2004 May.
Article in English | MEDLINE | ID: mdl-15078939

ABSTRACT

The erythroleukemia-inducing Friend spleen focus-forming virus (SFFV) encodes a unique envelope protein, gp55, which interacts with the erythropoietin (Epo) receptor complex, causing proliferation and differentiation of erythroid cells in the absence of Epo. Susceptibility to SFFV-induced erythroleukemia is conferred by the Fv-2 gene, which encodes a short form of the receptor tyrosine kinase Stk/Ron (sf-Stk) only in susceptible strains of mice. We recently demonstrated that sf-Stk becomes activated by forming a strong interaction with SFFV gp55. To examine the biological consequences of activated sf-Stk on erythroid cell growth, we prepared retroviral vectors which express sf-Stk, either in conjunction with gp55 or alone in a constitutively activated mutant form, and tested them for their ability to induce Epo-independent erythroid colonies ex vivo and disease in mice. Our data indicate that both gp55-activated sf-Stk and the constitutively activated mutant of sf-Stk induce erythroid cells from Fv-2-susceptible and Fv-2-resistant (sf-Stk null) mice to form Epo-independent colonies. Mutational analysis of sf-Stk indicated that a functional kinase domain and 8 of its 12 tyrosine residues are required for the induction of Epo-independent colonies. Further studies demonstrated that coexpression of SFFV gp55 with sf-Stk significantly extends the half-life of the kinase. When injected into Fv-2-resistant mice, neither the gp55-activated sf-Stk nor the constitutively activated mutant caused erythroleukemia. Surprisingly, both Fv-2-susceptible and -resistant mice injected with the gp55-sf-Stk vector developed clinical signs not previously associated with SFFV-induced disease. We conclude that sf-Stk, activated by either point mutation or interaction with SFFV gp55, is sufficient to induce Epo-independent erythroid colonies from both Fv-2-susceptible and -resistant mice but is unable to cause erythroleukemia in Fv-2-resistant mice.


Subject(s)
Erythroid Cells/metabolism , Point Mutation , Receptor Protein-Tyrosine Kinases/metabolism , Spleen Focus-Forming Viruses/pathogenicity , Viral Envelope Proteins/metabolism , Animals , Cell Line , Erythroid Cells/cytology , Erythropoietin/metabolism , Genetic Vectors , Leukemia, Erythroblastic, Acute/physiopathology , Leukemia, Erythroblastic, Acute/virology , Mice , Mice, Inbred C57BL , Receptor Protein-Tyrosine Kinases/genetics , Retroviridae Infections/physiopathology , Retroviridae Infections/virology , Spleen Focus-Forming Viruses/metabolism , Tumor Virus Infections/physiopathology , Tumor Virus Infections/virology
17.
J Virol ; 77(9): 5145-51, 2003 May.
Article in English | MEDLINE | ID: mdl-12692217

ABSTRACT

PVC-211 murine leukemia virus (MuLV) is a neuropathogenic variant of Friend MuLV (F-MuLV) which causes a rapidly progressive spongiform neurodegenerative disease in rodents. The primary target of PVC-211 MuLV infection in the brain is the brain capillary endothelial cell (BCEC), which is resistant to F-MuLV infection. Previous studies have shown that changes in the envelope gene of PVC-211 MuLV confer BCEC tropism to the virus. However, little is known about how infection of BCECs by PVC-211 MuLV induces neurological disease. Previous results suggest that nitric oxide (NO), which has been implicated as a potential neurotoxin, is involved in PVC-211 MuLV-induced neurodegeneration. In this study, we show that expression of inducible nitric oxide synthase (iNOS), which produces NO from L-arginine, is induced in BCECs from PVC-211 MuLV-infected rats. Furthermore, elevated levels of a 32-kDa cellular protein modified by 3-nitrotyrosine, which is a hallmark of NO production, were observed in virus-infected BCECs. BCECs from rats infected with BCEC-tropic but nonneuropathogenic PVF-e5 MuLV, which is a chimeric virus between PVC-211 MuLV and F-MuLV, fail to induce either iNOS expression or elevation of tyrosine nitration of a 32-kDa protein. These results suggest that expression of iNOS and nitration of tyrosine residues of a 32-kDa protein in PVC-211 MuLV-infected BCECs may play an important role in neurological disease induction.


Subject(s)
Brain/blood supply , Endothelium, Vascular/virology , Leukemia Virus, Murine/pathogenicity , Nitric Oxide Synthase/biosynthesis , Proteins/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , 3T3 Cells , Animals , Brain/enzymology , Brain/metabolism , Brain/virology , Capillaries/virology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Mice , Nervous System/pathology , Nervous System/virology , Nitric Oxide Synthase Type II , Rats , Rats, Inbred F344 , Retroviridae Infections/physiopathology , Retroviridae Infections/virology , Tumor Virus Infections/physiopathology , Tumor Virus Infections/virology
18.
J Biol Chem ; 278(15): 13520-30, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12574163

ABSTRACT

A novel protein kinase, polyploidy-associated protein kinase (PAPK), was isolated using a subtraction cDNA library approach from a mouse erythroleukemia cell line that had been induced to polyploidy after serum withdrawal. PAPK shares homology with members of the Ste20/germinal center kinase family of protein kinases and is ubiquitously expressed as two spliced forms, PAPK-A and PAPK-B, that encode for proteins of 418 and 189 amino acids, respectively. The expression of endogenous PAPK-A protein increased after growth factor withdrawal in murine hematopoietic and fibroblast cells. When tested in an in vitro kinase assay, PAPK-A was activated in response to the stress-inducing agent hydrogen peroxide and slightly by fetal calf serum. Biochemical characterization of the PAPK-A-initiated pathway revealed that this novel kinase does not affect MAP kinase activity but can stimulate both c-Jun N-terminal kinase 1 (JNK1) and ERK6/p38 gamma. The kinase activity of PAPK appears to be required for the activation of ERK6/p38 gamma but not JNK1. When an inducible construct of PAPK-A was expressed in stably transfected NIH3T3 cells, the cells exhibited distinct cytoskeletal changes and became resistant to apoptotic cell death induced by serum withdrawal, effects of PAPK that require its kinase activity. These data suggest that PAPK is a new member of the Ste20/germinal center kinase family that modulates cytoskeletal organization and cell survival.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Culture Media, Serum-Free , DNA, Complementary/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Leukemia, Erythroblastic, Acute , MAP Kinase Signaling System/genetics , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Polyploidy , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic , Tumor Cells, Cultured
19.
J Virol ; 76(3): 1527-32, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11773427

ABSTRACT

We have molecularly cloned a feline leukemia virus (FeLV) (clone 33) from a domestic cat with acute myeloid leukemia (AML). The long terminal repeat (LTR) of this virus, like the LTRs present in FeLV proviruses from other cats with AML, contains an unusual structure in its U3 region upstream of the enhancer (URE) consisting of three tandem direct repeats of 47 bp. To test the disease potential and specificity of this unique FeLV LTR, we replaced the U3 region of the LTR of the erythroleukemia-inducing Friend murine leukemia virus (F-MuLV) with that of FeLV clone 33. When the resulting virus, F33V, was injected into newborn mice, almost all of the mice eventually developed hematopoietic malignancies, with a significant percentage being in the myeloid lineage. This is in contrast to mice injected with an F-MuLV recombinant containing the U3 region of another FeLV that lacks repetitive URE sequences, none of which developed myeloid malignancies. Examination of tumor proviruses from F33V-infected mice failed to detect any changes in FeLV U3 sequences other than that in the URE. Like F-MuLV-infected mice, those infected with the F-MuLV/FeLV recombinants were able to generate and replicate mink cell focus-inducing viruses. Our studies are consistent with the idea that the presence of repetitive sequences upstream of the enhancer in the LTR of FeLV may favor the activation of this promoter in myeloid cells and contribute to the development of malignancies in this hematopoietic lineage.


Subject(s)
DNA, Viral , Friend murine leukemia virus/genetics , Leukemia Virus, Feline/genetics , Terminal Repeat Sequences , Animals , Base Sequence , Cats , Cell Line , Cloning, Molecular , Friend murine leukemia virus/physiology , Leukemia, Erythroblastic, Acute/virology , Leukemia, Experimental/virology , Leukemia, Myeloid/virology , Lymphoma/virology , Mice , Mink Cell Focus-Inducing Viruses/genetics , Molecular Sequence Data , Recombination, Genetic , Retroviridae , Retroviridae Infections/virology , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Tumor Virus Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...