Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
J Virol ; 75(24): 12439-45, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11711634

RESUMEN

Previous studies from our laboratory demonstrated that PVC-211 murine leukemia virus (MuLV), a neuropathogenic variant of Friend MuLV (F-MuLV), had undergone genetic changes which allowed it to efficiently infect rat brain capillary endothelial cells (BCEC) in vivo and in vitro. Two amino acid changes from F-MuLV in the putative receptor binding domain (RBD) of the envelope surface protein of PVC-211 MuLV (Glu-116 to Gly and Glu-129 to Lys) were shown to be sufficient for conferring BCEC tropism on PVC-211 MuLV. Recent examination of the unique RBD of PVC-211 MuLV revealed that the substitution of Lys for Glu at position 129 created a new heparin-binding domain that overlapped a heparin-binding domain common to ecotropic MuLVs. In this study we used heparin-Sepharose columns to demonstrate that PVC-211 MuLV, but not F-MuLV, can bind efficiently to heparin and that one or both of the amino acids in the RBD of PVC-211 MuLV that are associated with BCEC tropism are responsible. We further showed that heparin can enhance or inhibit MuLV infection and that the mode of action is dependent on heparin concentration, sulfation of heparin, and the affinity of the virus for heparin. Our results suggest that the amino acid changes that occurred in the envelope surface protein of PVC-211 MuLV may allow the virus to bind strongly to the surface of BCEC via heparin-like molecules, increasing the probability that the virus will bind to its cell surface receptor and efficiently infect these cells.


Asunto(s)
Encéfalo/virología , Endotelio Vascular/virología , Heparina/metabolismo , Virus de la Leucemia Murina/fisiología , Proteínas del Envoltorio Viral/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Capilares/virología , Heparina/farmacología , Virus de la Leucemia Murina/efectos de los fármacos , Datos de Secuencia Molecular , Ratas , Proteínas del Envoltorio Viral/fisiología
2.
J Virol ; 75(17): 7893-903, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11483734

RESUMEN

The Friend spleen focus-forming virus (SFFV) encodes a unique envelope glycoprotein, gp55, which allows erythroid cells to proliferate and differentiate in the absence of erythropoietin (Epo). SFFV gp55 has been shown to interact with the Epo receptor complex, causing constitutive activation of various signal-transducing molecules. When injected into adult mice, SFFV induces a rapid erythroleukemia, with susceptibility being determined by the host gene Fv-2, which was recently shown to be identical to the gene encoding the receptor tyrosine kinase Stk/Ron. Susceptible, but not resistant, mice encode not only full-length Stk but also a truncated form of the kinase, sf-Stk, which may mediate the biological effects of SFFV infection. To determine whether expression of SFFV gp55 leads to the activation of sf-Stk, we expressed sf-Stk, with or without SFFV gp55, in hematopoietic cells expressing the Epo receptor. Our data indicate that sf-Stk interacts with SFFV gp55 as well as gp55(P), the biologically active form of the viral glycoprotein, forming disulfide-linked complexes. This covalent interaction, as well as noncovalent interactions with SFFV gp55, results in constitutive tyrosine phosphorylation of sf-Stk and its association with multiple tyrosine-phosphorylated signal-transducing molecules. In contrast, neither Epo stimulation in the absence of SFFV gp55 expression nor expression of a mutant of SFFV that cannot interact with sf-Stk was able to induce tyrosine phosphorylation of sf-Stk or its association with any signal-transducing molecules. Covalent interaction of sf-Stk with SFFV gp55 and constitutive tyrosine phosphorylation of sf-Stk can also be detected in an erythroleukemia cell line derived from an SFFV-infected mouse. Our results suggest that SFFV gp55 may mediate its biological effects in vivo by interacting with and activating a truncated form of the receptor tyrosine kinase Stk.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/química , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Virus Formadores de Foco en el Bazo/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Leucemia Experimental/virología , Ratones , Mutación , Fosforilación , Plásmidos/genética , Receptores de Eritropoyetina/metabolismo , Infecciones por Retroviridae/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Virus Formadores de Foco en el Bazo/genética , Células Tumorales Cultivadas , Infecciones Tumorales por Virus/virología , Tirosina/metabolismo , Proteínas del Envoltorio Viral/genética
3.
Dev Biol (Basel) ; 106: 163-7; discussion 167-8, 253-63, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11761228

RESUMEN

Using neoplastic cell lines as substrates for vaccine development could inadvertently result in viral-viral or viral-cellular interactions whose biological consequences are unclear. In this review, the generation of mink cell focus-inducing (MCF) retroviruses in the mouse is discussed as a model for understanding how viral-viral and viral-cellular interactions can result in the generation of new retroviruses with pathological consequences.


Asunto(s)
Leucemia Experimental/virología , Fusión de Membrana , Virus Inductores de Focos en Células del Visón/fisiología , Modelos Biológicos , Animales , Ratones , Virus Inductores de Focos en Células del Visón/patogenicidad , Células Tumorales Cultivadas
4.
J Immunol ; 165(11): 6372-80, 2000 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11086075

RESUMEN

The ubiquitin-dependent proteasome-mediated (Ub-Pr) degradation pathway has been shown to regulate a large variety of substrates, including nuclear, cytosolic, and membrane proteins. In mammalian systems, polyubiquitin modification has been identified in a number of cell surface receptors for more than a decade; however, its biological significance has remained unclear until recently. For growth factor receptors with intrinsic tyrosine kinase domains, polyubiquitination is believed to trigger the internalization and subsequent degradation via the lysosomal pathway. In this study we provide the first evidence that non-tyrosine kinase-type cytokine surface receptors, IL-9R alpha-chain, IL-2 receptor ss-chain, and erythropoietin receptor, can be polyubiquitinated and degraded by proteasomes. The Ub-Pr pathway regulates both the basal level turnover and the ligand-induced degradation of the receptors. A previously identified putative molecular chaperon, valosin-containing protein, undergoes tyrosine phosphorylation in a cytokine-dependent manner and associates with the receptor complexes following receptor engagement, suggesting that valosin-containing protein may target the ubiquitinated receptors to the proteasome for degradation.


Asunto(s)
Biopolímeros/fisiología , Cisteína Endopeptidasas/fisiología , Eritropoyetina/metabolismo , Interleucina-2/metabolismo , Interleucina-9/metabolismo , Complejos Multienzimáticos/fisiología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal/inmunología , Ubiquitinas/fisiología , Adenosina Trifosfatasas , Animales , Biopolímeros/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Células Clonales , Eritropoyetina/fisiología , Interleucina-2/fisiología , Ratones , Chaperonas Moleculares/fisiología , Fosforilación , Poliubiquitina , Complejo de la Endopetidasa Proteasomal , Receptores de Eritropoyetina/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-2/metabolismo , Receptores de Interleucina-9 , Linfocitos T/enzimología , Linfocitos T/metabolismo , Células Tumorales Cultivadas , Tirosina/metabolismo , Ubiquitinas/metabolismo , Proteína que Contiene Valosina
5.
J Virol ; 74(18): 8444-51, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10954544

RESUMEN

Interaction of erythropoietin (Epo) with its cell surface receptor activates signal transduction pathways which result in the proliferation and differentiation of erythroid cells. Infection of erythroid cells with the Friend spleen focus-forming virus (SFFV) leads to the interaction of the viral envelope glycoprotein with the Epo receptor and renders these cells Epo independent. We previously reported that SFFV induces Epo independence by constitutively activating components of several Epo signal transduction pathways, including the Jak-Stat and the Raf-1/mitogen-activated protein kinase (MAPK) pathways. To further evaluate the mechanism by which SFFV activates the Raf-1/MAPK pathway, we investigated the effects of SFFV on upstream components of this pathway, and our results indicate that SFFV activates Shc and Grb2 and that this leads to Ras activation. While studies with a dominant-negative Ras indicated that Ras was required for Epo-induced proliferation of normal erythroid cells, the Epo-independent growth of SFFV-infected cells can still occur in the absence of Ras, although at reduced levels. In contrast, protein kinase C (PKC) was shown to be required for the Epo-independent proliferation of SFFV-infected cells. Further studies indicated that PKC, which is thought to be involved in the activation of both Raf-1 and MAPK, was required only for the activation of MAPK, not Raf-1, in SFFV-infected cells. Our results indicate that Ras and PKC define two distinct signals converging on MAPK in both Epo-stimulated and SFFV-infected erythroid cells and that activation of only PKC is sufficient for the Epo-independent proliferation of SFFV-infected cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular , Eritroblastos/metabolismo , Eritropoyetina/metabolismo , Guanosina Trifosfato/metabolismo , Proteína Quinasa C/metabolismo , Virus Formadores de Foco en el Bazo/metabolismo , Sulfonamidas , Proteínas ras/metabolismo , Western Blotting , División Celular , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Eritroblastos/virología , Proteína Adaptadora GRB2 , Humanos , Indoles/farmacología , Isoquinolinas/farmacología , Maleimidas/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Unión Proteica , Proteína Quinasa C/antagonistas & inhibidores , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Estaurosporina/farmacología , Transfección , Células Tumorales Cultivadas
6.
J Virol ; 74(7): 3037-45, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10708418

RESUMEN

The erythroleukemia-inducing Friend spleen focus-forming virus (SFFV) encodes a unique envelope glycoprotein which allows erythroid cells to proliferate and differentiate in the absence of erythropoietin (Epo). In an effort to understand how SFFV causes Epo independence, we have been examining erythroid cells rendered factor independent by SFFV infection for constitutive activation of signal-transducing molecules. Previous studies from our laboratory showed that various signal-transducing molecules known to be activated by Epo, including Stat proteins and components of the Raf-1/MAP kinase pathway, are constitutively activated in SFFV-infected erythroid cells in the absence of Epo. Since another signal transduction pathway involving activation of phosphatidylinositol 3-kinase (PI 3-kinase) after Epo stimulation plays an important role in erythroid cell proliferation and differentiation, we carried out studies to determine if this pathway was also activated in SFFV-infected cells in the absence of Epo. Our studies show that PI 3-kinase is constitutively activated in erythroid cells rendered factor independent by infection with SFFV and that PI 3-kinase activity, but not Epo receptor tyrosine phosphorylation, is required for the proliferation of these cells in the absence of Epo. We further show that in SFFV-infected erythroid cells grown in the absence of Epo, PI 3-kinase associates with the insulin receptor substrate (IRS)-related adapter molecules IRS-2, Gab1, and Gab2, which are constitutively tyrosine phosphorylated in SFFV-infected cells. Finally, Akt, a protein kinase that is one of the downstream effectors of PI 3-kinase, and SHIP, a lipid phosphatase that is important for Akt activation through PI 3-kinase, are both tyrosine phosphorylated in SFFV-infected cells grown in the absence of Epo. Our results indicate that induction of Epo independence by SFFV requires the activation of PI 3-kinase and suggest that constitutive activation of this kinase in SFFV-infected cells may occur primarily through interaction of PI 3-kinase with constitutively phosphorylated IRS-related adapter molecules.


Asunto(s)
Eritrocitos/virología , Eritropoyetina/fisiología , Virus de la Leucemia Murina de Friend/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Activación Enzimática , Proteína Oncogénica v-akt , Fosforilación , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/metabolismo , Células Tumorales Cultivadas , Tirosina/metabolismo
7.
Int J Biochem Cell Biol ; 31(10): 1089-109, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10582341

RESUMEN

The proliferation and differentiation of erythroid cells is a highly regulated process that is controlled primarily at the level of interaction of erythropoietin (Epo) with its specific cell surface receptor (EpoR). However, this process is deregulated in mice infected with the Friend spleen focus-forming virus (SFFV). Unlike normal erythroid cells, erythroid cells from SFFV-infected mice are able to proliferate and differentiate in the absence of Epo, resulting in erythroid hyperplasia and leukemia. Over the past 20 years, studies have been carried out to identify the viral genes responsible for the pathogenicity of SFFV and to understand how expression of these genes leads to the deregulation of erythropoiesis in infected animals. The studies have revealed that SFFV encodes a unique envelope glycoprotein which interacts specifically with the EpoR at the cell surface, resulting in activation of the receptor and subsequent activation of erythroid signal transduction pathways. This leads to the proliferation and differentiation of erythroid precursor cells in the absence of Epo. Although the precise mechanism by which the viral protein activates the EpoR is not yet known, it has been proposed that it causes dimerization of the receptor, resulting in constitutive activation of Epo signal transduction pathways. While interaction of the SFFV envelope glycoprotein with the EpoR leads to Epo-independent erythroid hyperplasia, this is not sufficient to transform these cells. Transformation requires the viral activation of the cellular gene Sfpi-1, whose product is thought to block erythroid cell differentiation. By understanding how SFFV can deregulate erythropoiesis, we may gain insights into the causes and treatment of related diseases in man.


Asunto(s)
Eritropoyesis , Leucemia Eritroblástica Aguda/fisiopatología , Leucemia Eritroblástica Aguda/virología , Virus Formadores de Foco en el Bazo/fisiología , Animales , Transformación Celular Viral , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/virología , Humanos , Hiperplasia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiología , Ratones , Receptores de Eritropoyetina/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/fisiología
8.
J Virol ; 73(10): 8623-9, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10482615

RESUMEN

Entry of ecotropic murine leukemia virus (MuLV) into host cells is initiated by interaction between the receptor-binding domain of the viral SU protein and the third extracellular domain (TED) of the receptor, cationic amino acid transporter 1 (CAT1). To study the molecular basis for the retrovirus-receptor interaction, mouse CAT1 (mCAT1) was expressed in human 293 cells as a fusion protein with jellyfish green fluorescent protein (GFP). Easily detected by fluorescence microscopy and immunoblot analysis with anti-GFP antibodies, the mCAT1-GFP fusion protein was expressed in an N-glycosylated form on the cell surface and in the Golgi apparatus, retaining the ecotropic receptor function. The system was applied to compare Friend MuLV (F-MuLV) and its neuropathogenic variant, PVC-211 MuLV, which exhibits a unique cellular tropism and host range, for the ability to use various CAT family members as a receptor. The results indicated that F-MuLV and PVC-211 MuLV could infect the cells expressing wild-type mCAT1 at comparable efficiencies and that rat CAT3, but not mCAT2, conferred a low but detectable level of susceptibility to F-MuLV and PVC-211 MuLV. The data also suggested that CAT proteins might be expressed in an oligomeric form. Further application of the system developed in this study may provide useful insights into the entry mechanism of ecotropic MuLV.


Asunto(s)
Proteínas Portadoras/fisiología , Virus de la Leucemia Murina/fisiología , Proteínas de la Membrana/fisiología , Receptores Virales/fisiología , Replicación Viral , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos Básicos , Animales , Gatos , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes , Ratones , Datos de Secuencia Molecular , Ratas , Alineación de Secuencia
9.
J Virol ; 72(4): 3423-6, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9525674

RESUMEN

PVC-441 murine leukemia virus (MuLV) is a member of the PVC group of Friend MuLV (F-MuLV)-derived neuropathogenic retroviruses. In order to determine the molecular basis for the difference in neuropathogenicity between PVC-441 and the previously characterized PVC-211 MuLVs, the entire nucleotide sequence of PVC-441 MuLV was determined and compared with those of PVC-211 and F-MuLV. The results suggest that PVC-441 and PVC-211 MuLVs were formed as a result of random mutations of F-MuLV and developed differently. The distinct pathogenicities of PVC-441 and PVC-211 MuLVs were maintained in the viruses regenerated from their molecular clones, and the sequences responsible for the pathological differences observed can be localized to the env gene. The amino acid sequence of PVC-441 deduced from its nucleotide sequence revealed a number of differences from PVC-211, the most striking of which was a difference at position 129 of the SU proteins in the two viruses. Host range studies with a brain capillary endothelial cell line (RTEC-6) and Chinese hamster ovary cells (CHO-K1) revealed that PVC-441, like PVC-211, could infect these cells but its efficiency of infection was lower than that of PVC-211. These results may account for the difference in neuropathogenicity between PVC-441 and PVC-211.


Asunto(s)
Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina/genética , Animales , Secuencia de Bases , Células CHO , Cricetinae , ADN Viral , Virus de la Leucemia Murina de Friend/patogenicidad , Productos del Gen env/genética , Productos del Gen env/fisiología , Genes gag , Genes pol , Virus de la Leucemia Murina/patogenicidad , Leucemia Experimental/virología , Ratones , Datos de Secuencia Molecular , Parálisis/virología , Ratas , Ratas Endogámicas F344 , Infecciones por Retroviridae/virología , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Infecciones Tumorales por Virus/virología
10.
J Virol ; 72(2): 919-25, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9444983

RESUMEN

The erythroleukemia-inducing Friend spleen focus-forming virus (SFFV) encodes a unique envelope glycoprotein which allows erythroid cells to proliferate and differentiate in the absence of erythropoietin (Epo). In an attempt to understand how the virus causes Epo independence, we have been studying signal transduction pathways activated by Epo to determine if SFFV exerts its biological effects by constitutively activating any of these pathways in the absence of Epo. We previously demonstrated that Stat proteins, the downstream components of the Epo-induced Jak-Stat pathway, are constitutively activated in SFFV-infected cells. In this study, we demonstrate that SFFV also activates Raf-1, MEK and mitogen-activated protein (MAP) kinase, the downstream components of the Raf-1/MAP kinase pathway. This pathway was activated in cells infected with the polycythemia-inducing strain of SFFV, which induces both proliferation and differentiation of erythroid cells in the absence of Epo, as well as in cells infected with the anemia-inducing strain of the virus, which still require Epo for differentiation. Inhibition of Raf-1 by using antisense oligonucleotides led to a partial inhibition of the Epo-independent proliferation of SFFV-infected cells. Expression of the transcription factors c-Jun and JunB, but not c-Fos, was induced in SFFV-infected cells in the absence of Epo, suggesting that constitutive activation of the Raf-1/MAP kinase pathway by the virus may result in deregulation of AP-1 activity. We conclude from our studies that infection of erythroid cells with SFFV leads to the constitutive activation of signal transduction molecules in both the Jak-Stat and Raf-1/MAP kinase pathways and that both of these pathways must be activated to achieve maximum proliferation and differentiation of erythroid cells in the absence of Epo.


Asunto(s)
Leucemia Experimental/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Infecciones por Retroviridae/metabolismo , Transducción de Señal , Virus Formadores de Foco en el Bazo , Infecciones Tumorales por Virus/metabolismo , Anemia/virología , Animales , Activación Enzimática , Ratones , Policitemia/virología
11.
J Virol ; 71(8): 6168-73, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9223511

RESUMEN

PVC-211 murine leukemia virus (MuLV) causes neurodegenerative disease following inoculation of neonatal, but not adult, mice and rats. It was previously shown that tropism for brain capillary endothelial cells (CEC) was a determinant of the viral neuropathogenicity. In this study, we demonstrate that host age-dependent replication of PVC-211 MuLV in vivo occurs in CEC in the brain as well as in other organs, such as the liver, kidney, and heart. In contrast, primary explant cultures of CEC derived from brains and livers of adult and neonatal rats could be infected by PVC-211 MuLV, suggesting that the age-dependent susceptibility was abrogated in vitro. Although CEC were generally less susceptible to MuLV-mediated gene transduction than fibroblasts, treatment of CEC with 2-deoxyglucose followed by inoculation of a PVC-211 MuLV-pseudotyped vector in the absence of heparin improved the transduction efficiency. These observations support the possibility that PVC-211 MuLV may be useful for establishing models of CEC gene transduction.


Asunto(s)
Endotelio Vascular/virología , Virus de la Leucemia Murina de Friend/fisiología , Técnicas de Transferencia de Gen , Animales , Encéfalo/virología , Capilares/virología , Células Cultivadas , Desoxiglucosa/farmacología , Hígado/virología , Ratas , Ratas Endogámicas F344
12.
Leukemia ; 11 Suppl 3: 233-5, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9209351

RESUMEN

A neuropathogenic variant of Friend MuLV, PVC-211, causes rapidly progressive spongiform neurodegeneration in susceptible rats and mice. Major targets of PVC-211 MuLV infection are brain capillary endothelial cells (BCEC), suggesting that virus-infected BCEC may play crucial roles in neurological disease induction. Consistent with this possibility, studies using chimeric viruses constructed between PVC-211 MuLV and non-neuropathogenic Friend MuLV have revealed that the BCEC tropism of the virus correlates with its neuropathogenicity. Possible involvement of cytokine expression by PVC-211 MuLV-infected BCEC in the induction of neuropathological changes will be discussed.


Asunto(s)
Encéfalo/virología , Circulación Cerebrovascular , Endotelio Vascular/virología , Virus de la Leucemia Murina de Friend/patogenicidad , Animales , Encéfalo/patología , Capilares , Células Cultivadas , Quimera , Virus de la Leucemia Murina de Friend/genética , Ratones , Ratas , Mapeo Restrictivo
13.
Leukemia ; 11 Suppl 3: 251-4, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9209356

RESUMEN

The erythroleukemia-inducing Friend spleen focus-forming virus (SFFV) encodes a unique envelope glycoprotein which allows erythroid cells to proliferate and differentiate in the absence of the erythroid hormone erythropoietin (Epo). In an attempt to understand how the virus alters the growth of erythroid cells, studies were carried out to determine if virus infection leads to the constitutive activation of the Jak-Stat pathway, one of the signal transduction pathways activated by Epo. Our data indicates that expression of SFFV in erythroid cells leads to the constitutive activation of the same Stat proteins that are transiently activated by Epo. While constitutive activation of Stat proteins by SFFV is associated with Epo-independent proliferation of splenic erythroid progenitor cells from Fv-2-sensitive mice and Epo-dependent HCD-57 cells, it is not sufficient to induce their differentiation. Although constitutive activation of the same Stat proteins is detected in erythroid cells from SFFV-infected Fv-2-resistant mice, it does not lead to their Epo-independent growth. It is also not required for transformation of erythroid cells by SFFV. Studies are in progress to identify the mechanism by which Stat proteins are phosphorylated in SFFV-infected cells in the absence of Epo. Although it has been shown that Epo activates Stat proteins through Jak2 kinase, our results suggest that the SFFV-induced Stat protein activation is Jak2-independent.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Genes fos , Células Madre Hematopoyéticas/fisiología , Leucemia Eritroblástica Aguda/fisiopatología , Transducción de Señal/fisiología , Virus Formadores de Foco en el Bazo/fisiología , Replicación Viral , Animales , Diferenciación Celular , Línea Celular , Eritropoyesis , Eritropoyetina/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/virología , Leucemia Eritroblástica Aguda/virología , Ratones , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Replicación Viral/efectos de los fármacos
14.
J Virol ; 70(12): 8534-9, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8970977

RESUMEN

PVC-211 murine leukemia virus (MuLV) is a neuropathogenic variant of Friend MuLV (F-MuLV). Previous studies from our laboratory demonstrated that unlike the parental F-MuLV, PVC-211 MuLV can infect rat brain capillary endothelial cells efficiently and that it has acquired genetic changes responsible for its expanded cellular tropism. To determine if PVC-211 MuLV also has expanded its host range, we tested its infectivity on Chinese hamster ovary-derived CHO-K1 cells, which are generally resistant to ecotropic MuLV. The results indicated that PVC-211 MuLV, but not F-MuLV, was highly infectious for CHO-K1 cells. Studies using glycosylation inhibitors and glycosylation mutants of CHO-K1 cells, as well as interference studies, suggested that PVC-211 MuLV has acquired the ability to interact with the ecotropic MuLV receptor on CHO-K1 cells that has undergone glycosylation-dependent modification. Using chimeric viruses between PVC-211 MuLV and F-MuLV, we were able to localize the viral genetic element crucial for CHO-K1 cell tropism within the env gene of PVC-211 MuLV and show that glycine at position 116 and lysine at position 129 of the envelope glycoprotein SU were important. These viral determinants also appear to confer tropism for other hamster cells resistant to ordinary ecotropic MuLVs. Further studies on the interaction between PVC-211 MuLV and the receptor on hamster cells may provide novel insights into the molecular mechanisms for receptor recognition and binding by viral envelope glycoproteins.


Asunto(s)
Virus de la Leucemia Murina de Friend/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células CHO , Línea Celular , Cricetinae , ADN Viral , ADN-Citosina Metilasas/metabolismo , Desoxiglucosa/farmacología , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Virus de la Leucemia Murina de Friend/genética , Productos del Gen env/genética , Glicosilación/efectos de los fármacos , Virus de la Leucemia del Gibón/metabolismo , Ratones , Datos de Secuencia Molecular , Ratas , Receptores Virales/metabolismo , Tunicamicina/farmacología
15.
Oncogene ; 13(5): 1037-42, 1996 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-8806693

RESUMEN

The myb-ets-containing ME26 virus causes erythroleukemia in mice by a novel mechanism involving the inappropriate activation of erythroid-specific genes in hematopoietic precursor cells. We have previously shown that the ME26 viral protein can transactivate the GATA-1 promoter in transient transactivation assays carried out in mouse fibroblasts. The mouse GATA-1 promoter, whose activity is regulated by the GATA-1 protein itself, contains a double GATA consensus sequence at its 5' end and two CACCC elements at its 3' end, both of which are crucial for promoter activity in erythroid cells, as well as a nonconsensus GATA sequence and several putative c-myb and c-ets binding sites. In order to determine which sequences in the GATA-1 promoter are crucial for activation by the ME26 viral protein, we made deletions of the promoter, cloned them into a luciferase expression vector and tested their activity in mouse fibroblasts, which do not express GATA-1. Our results indicate that sequences in the 3' end of the GATA-1 promoter, which include two CACCC elements, are essential for transactivation by ME26 virus, while other upstream sites contribute to full activation by the virus. Mutation of the CACCC sites abolishes ME26 viral transactivation. The interaction of cell extracts containing ME26 viral protein and the GATA-1 promoter fragment containing the two CACCC elements was examined by electrophoretic mobility shift analysis (EMSA) and the results showed no direct interaction between the two. However, we could detect the ubiquitous transcription factor Sp1 bound to this sequence. These data demonstrate that the CACCC element is necessary for GATA-1 promoter transactivation by ME26 virus and that the viral protein may indirectly transactivate the promoter by binding to Sp1.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Oncogénicas de Retroviridae/genética , Retroviridae/química , Transactivadores , Transactivadores/genética , Factores de Transcripción/genética , Células 3T3/virología , Animales , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Ratones , Mutación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myb , Retroviridae/genética , Proteínas Oncogénicas de Retroviridae/metabolismo , Eliminación de Secuencia , Factor de Transcripción Sp1/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
16.
Virology ; 215(2): 142-51, 1996 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8560761

RESUMEN

PVC-211 murine leukemia virus (MuLV) is a neuropathogenic variant of Friend MuLV (F-MuLV) that causes a rapidly progressive neurodegenerative disease in susceptible rodents. PVC-211 MuLV, but not the parental F-MuLV, can infect rat brain capillary endothelial cells (BCEC) efficiently, and the major determinant for BCEC tropism of PVC-211 MuLV is localized within the XbaI-BamHI fragment of the viral genome containing the 5' half of the env gene. To further dissect the XbaI-BamHI region for its effects on BCEC tropism, we constructed recombinant viruses between PVC-211 MuLV and F-MuLV and tested their infectivity on a cell line established from rat BCEC. Our results indicated that Glu116-to-Gly and Glu129-to-Lys substitutions in the background of the F-MuLV envelope SU protein were sufficient for conferring BCEC tropism on the virus. Interference studies of these viruses on Rat-1 fibroblastic cells showed that the structure of the SU protein encoded by the XbaI-BamHI region also has significant effects on their affinity for the rat ecotropic MuLV receptor. These results support the possibility that structural elements I and II of the SU protein are important determinants for virus-receptor interaction.


Asunto(s)
Encéfalo/virología , Endotelio Vascular/virología , Virus de la Leucemia Murina de Friend/fisiología , Proteínas Oncogénicas de Retroviridae/fisiología , Proteínas del Envoltorio Viral/fisiología , Interferencia Viral , Células 3T3 , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , ADN Viral , Fibroblastos/citología , Virus de la Leucemia Murina de Friend/química , Virus de la Leucemia Murina de Friend/genética , Vectores Genéticos , Ratones , Datos de Secuencia Molecular , Provirus/genética , Ratas , Receptores Virales/metabolismo , Recombinación Genética , Proteínas Oncogénicas de Retroviridae/química , Proteínas Oncogénicas de Retroviridae/genética , Relación Estructura-Actividad , Transducción Genética , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Integración Viral
18.
Blood ; 85(6): 1454-62, 1995 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-7888668

RESUMEN

The signal transduction mechanism of erythropoietin (Epo), which regulates growth and differentiation of erythroid cells, is still unclear. Recent studies showing the activation by various ligands of a group of proteins called Stat (signal transducers and activators of transcription) proteins raised the possibility that such proteins may also be involved in the Epo signal transduction pathway. In this report, we show that Epo induces factors that specifically bind to the sis-inducible element and the gamma response region of the Fc gamma receptor factor I gene in the Epo-dependent mouse erythroleukemia cell line HCD-57. These factors contain phosphotyrosine and antibodies against Stat1 and Stat3 proteins reacted with them. In HCD-57 cells infected with Friend spleen focus-forming virus, which now grow in an Epo-independent manner, the DNA-binding factors were constitutively activated even in the absence of Epo. These results suggest that the factors induced by Epo contain components identical or related to known Stat proteins. It is also suggested that continuous activation of these DNA-binding factors may be responsible for the ability of spleen focus-forming virus to abrogate the Epo-dependence of HCD-57 cells and cause erythroleukemia in susceptible mice.


Asunto(s)
ADN/metabolismo , Eritropoyetina/farmacología , Virus Formadores de Foco en el Bazo/fisiología , Animales , Secuencia de Bases , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Datos de Secuencia Molecular , ARN Mensajero/análisis , Receptores de IgG/genética , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Transactivadores/genética , Transactivadores/fisiología , Células Tumorales Cultivadas
19.
Baillieres Clin Haematol ; 8(1): 225-47, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7663048

RESUMEN

The Friend spleen focus-forming virus has been a valuable tool for understanding the molecular events involved in the multiple stages of leukaemia. As summarized in Figure 3, the primary effect of SFFV, which occurs within days, is to cause a polyclonal proliferation of erythroid precursor cells that can proliferate in the absence of their normal regulator erythropoietin. This is the direct result of the unique envelope glycoprotein encoded by SFFV, which is transported to the cell surface and apparently interacts with the EpoR or another component of the multimeric EpoR complex, resulting in the constitutive activation of the Epo signal transduction pathway. Within this proliferating population of erythroid cells is a rare cell that has undergone several genetic changes due to the integration of the viral genome in specific sites in the mouse DNA. This leads to the activation of a gene encoding the PU.1 transcription factor, whose high expression in erythroid cells may be the cause of the block in differentiation that is characteristic of SFFV-transformed erythroid cells. SFFV integration can also lead to the inactivation of the p53 tumour supressor gene, giving these cells a growth advantage in the mouse. The disease induced by SFFV in mice is very similar to polycythaemia vera in humans (Golde et al, 1981). The major clinical feature of polycythaemia vera is the continuous expansion of the number of mature red blood cells in the presence of low serum Epo levels. Also, BFU-E and CFU-E from these patients can form in the absence of Epo like the analogous cells from SFFV-infected mice (Casadevall et al, 1982). It is possible that haematopoietic cells from individuals suffering from this disease express a protein similar to the envelope glycoprotein of SFFV that can interact with the EpoR and lead to its constitutive activation. Alternatively, these patients may contain a mutant EpoR gene that is constitutively activated like the mutant EpoR described earlier. As we understand more fully how the SFFV envelope protein constitutively activates te EpoR complex, we can begin to design therapies to counteract its action that can then be applied to treating patients with polycythaemia vera or other human diseases associated with uncontrolled erythropoiesis.


Asunto(s)
Virus Defectuosos/patogenicidad , Virus Helper/fisiología , Leucemia Eritroblástica Aguda/virología , Leucemia Experimental/virología , Infecciones por Retroviridae/virología , Virus Formadores de Foco en el Bazo/patogenicidad , Infecciones Tumorales por Virus/virología , Animales , Transformación Celular Neoplásica , Transformación Celular Viral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Virus Defectuosos/genética , Virus Defectuosos/fisiología , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/virología , Eritropoyesis , Eritropoyetina/fisiología , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/fisiología , Genes env , Genoma Viral , Virus Helper/genética , Hiperplasia , Ratones , Mutagénesis Insercional , Receptores de Eritropoyetina/fisiología , Proteínas Oncogénicas de Retroviridae , Transducción de Señal , Virus Formadores de Foco en el Bazo/genética , Virus Formadores de Foco en el Bazo/fisiología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/fisiología , Replicación Viral
20.
J Immunol ; 153(3): 1004-14, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-7913108

RESUMEN

Severe combined immune deficiency (SCID) mice have a defect in their recombinase system and cannot productively rearrange their immune receptor genes. Thus, SCID thymocytes are arrested at the immature "triple negative" phase, not expressing CD3, CD4, or CD8 surface markers. Whole body irradiation of SCID mice induced maturation of their thymocytes to the CD4+/CD8+ double positive, CD3+low stage of differentiation, and resulted in the generation of a thymic cortical region on histologic examination. No mature single positive T cells were detected in the thymus or the periphery. VDJ rearrangements of TCR-beta with restricted clonality were observed in the double positive cells from a given individual. The CD3 complex was expressed on some of these cells, but the cells failed to mobilize intracellular calcium after cross-linking with CD3 Abs. The double positive cells appeared several weeks after irradiation, persisted for many months in the thymus, and by 6 mo generally developed into metastatic lymphoma. Retroviral activation was undetectable in both the preneoplastic and transformed thymocytes. Thus, it appears that the earliest steps in T cell development can be induced in SCID mice by inducing DNA breaks with radiation. This system represents a model of early thymic development, preneoplasia, and neoplasia.


Asunto(s)
Ratones SCID/inmunología , Subgrupos de Linfocitos T/inmunología , Timoma/etiología , Timo/citología , Animales , Apoptosis , Complejo CD3/análisis , Linfocitos T CD4-Positivos/citología , Antígenos CD8/análisis , Calcio/metabolismo , Diferenciación Celular , Rayos gamma , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Ratones , Ratones Endogámicos BALB C , Subgrupos de Linfocitos T/citología , Timo/microbiología , Timo/patología , Factores de Tiempo , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA