Detection of Tumorigenic Cells in Friend Virus-Infected Mice: An In Vivo Methodological Investigation2, 3

1981 ◽  
Vol 66 (6) ◽  
pp. 1121-1127 ◽  
Author(s):  
Françoise Moreau-Gachelin ◽  
Françoise Wendling ◽  
Paule Bucau-Varlet ◽  
Martine Charon ◽  
Pierre Tambourin
Blood ◽  
1988 ◽  
Vol 71 (3) ◽  
pp. 800-803
Author(s):  
RA Fleischman

Abstract The leukemias induced by the Friend polycythemia virus and other leukemogenic retroviruses have previously not been transplantable until weeks or months after virus inoculation. Because tumor-specific immune mechanisms persist in both irradiated and nude mice, it has not been possible to determine if this result is due to rejection of cells already immortalized by retrovirus infection, or reflects an inherent limitation in the proliferative capacity and malignancy of these “preleukemic” cells. To clarify these issues, we have transplanted virus-infected bone marrow into mouse fetuses that are immunologically immature and thus incapable of graft rejection. We report here that within days of virus inoculation, transplantable cells capable of disease progression in certain fetal hosts can be detected with this technique. These results demonstrate that cells with the capacity for extensive leukemic proliferation arise very early in Friend virus- induced disease. However, successful transplantation was seen only in genetically anemic recipients (Wx/Wv), which are deficient in hematopoietic stem cells, and not in their normal littermates. Thus, in accord with recent in vitro observations, this in vivo data suggests that normal hematopoietic cells, independent of immune mechanisms, can suppress the malignant progression of transformed cells.


1979 ◽  
Vol 161 (2) ◽  
pp. 210-215 ◽  
Author(s):  
R. B. Raikow ◽  
R. F. Meredith ◽  
B. J. Brozovich ◽  
P. R. Seeman ◽  
A. E. Livingston ◽  
...  

1975 ◽  
Vol 142 (4) ◽  
pp. 936-948 ◽  
Author(s):  
R J Eckner

Exposure of NIH Swiss mouse embryo fibroblasts (MEF) to infectious Friend virus (FV) complex [containing defective spleen focus-forming virus (SFFV) and endogenous NB-tropic leukemia-inducing helper virus (LLV-F)] resulted in the productive infection of these cells by both SFFV and LLV-F. Stocks of SFFV derived after extensive growth in this Swiss MEF cell culture system are fully leukemogenic in adult mice for the induction of erythroleukemia and spleen foci. In addition, in vitro-derived LLV-F, when isolated free of SFFV, is fully leukemogenic for the induction of lymphatic leukemia when inoculated into susceptible newborn BALB/c mice. The host range of in vitro-derived FV complex (i.e., FV-TC) for focus formation in vivo is NB-tropic. Unlike in vivo-derived FV complex, FV-TC does not suppress the responsiveness of murine thymocytes to concanavalin A (Con A) in vitro. Rather, FV-TC acts as a mitogen to nonspecifically stimulate the proliferation of BALB/c thymocytes. The mitogenicity of in vitro-derived FV complex is directly associated with the presence of type-C virus particles, is a heat-labile and UV-sensitive property of the virus, and may be primarily due to LLV since equivalent amounts of LLV with or without SFFV present are equally mitogenic. One in vivo passage of FV-TC resulted in the total loss of this mitogenic property with the reappearance of full immunosuppressive properties. This result demonstrates a clear association between in vivo growth of FV and its ability to suppress mouse thymocytes, and suggests that FV complex (SFFV-LLV) is not inherently immunosuppressive for these cells. While the mechanism of this interconversion between immunostimulatory and fully suppressive virus is unknown, both virus markers appear to be dependent upon the presence of infectious FV.


1971 ◽  
Vol 8 (2) ◽  
pp. 304-309 ◽  
Author(s):  
A. Howard Fieldsteel ◽  
Peter J. Dawson ◽  
Carole Kurahara

2001 ◽  
Vol 21 (1) ◽  
pp. 73-80 ◽  
Author(s):  
You-Jun Li ◽  
Rachel R. Higgins ◽  
Brian J. Pak ◽  
Ramesh A. Shivdasani ◽  
Paul A. Ney ◽  
...  

ABSTRACT In previous studies, we identified a common site of retroviral integration designated Fli-2 in Friend murine leukemia virus (F-MuLV)-induced erythroleukemia cell lines. Insertion of F-MuLV at the Fli-2 locus, which was associated with the loss of the second allele, resulted in the inactivation of the erythroid cell- and megakaryocyte-specific genep45 NFE2 . Frequent disruption ofp45 NFE2 due to proviral insertion suggests a role for this transcription factor in the progression of Friend virus-induced erythroleukemias. To assess this possibility, erythroleukemia was induced by F-MuLV inp45 NFE2 mutant mice. Sincep45 NFE2 homozygous mice mostly die at birth, erythroleukemia was induced in +/− and +/+ mice. We demonstrate that +/− mice succumb to the disease moderately but significantly faster than +/+ mice. In addition, the spleens of +/− mice were significantly larger than those of +/+ mice. Of the 37 tumors generated from the +/− and +/+ mice, 10 gave rise to cell lines, all of which were derived from +/− mice. Establishment in culture was associated with the loss of the remaining wild-typep45 NFE2 allele in 9 of 10 of these cell lines. The loss of a functional p45NFE2 in these cell lines was associated with a marked reduction in globin gene expression. Expression of wild-typep45 NFE2 in the nonproducer erythroleukemic cells resulted in reduced cell growth and restored the expression of globin genes. Similarly, the expression ofp45 NFE2 in these cells also slows tumor growth in vivo. These results indicate thatp45 NFE2 functions as an inhibitor of erythroid cell growth and that perturbation of its expression contributes to the progression of Friend erythroleukemia.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 465-465
Author(s):  
Shuang Ni ◽  
Jie Xu ◽  
Hami Teal ◽  
Gen-Sheng Feng ◽  
Robert F. Paulson ◽  
...  

Abstract Friend Virus induced acute erythroleukemia provides an experimental system to study the multi-stage events involved in the pathogenesis of hematologic neoplasias. Friend erythroleukemia progresses in a biphasic manner: the initial phase is characterized by the polyclonal expansion of infected erythroid precursor cells, which is promoted by the activation of a naturally-occuring truncated form of the Stk receptor called short-form stk (Sf-Stk); and the later phase of the disease is marked by the acquisition of fully transformed erythroid leukemic cells in the spleen, blood, bone marrow and liver, leading to the development of erythroleukemia. Studies from our lab have demonstrated the importance of the kinase activity of Sf-Stk, its Grb2 binding site and recruitment of a Grb2/Gab2 complex in the development of Friend Virus induced erythroleukemia. In addition, we have shown that fusion proteins of Sf-Stk and Gab2 (Sf-Stk/Gab2), in which the C-terminal docking site tyrosines of Sf-Stk were replaced with the coding sequence of Gab2, are able to support Epo-independent colony formation. Here we demonstrate that Gab2−/− mice are partially resistant to Friend erythroleukemia in vivo and Gab2−/− erythroblasts fail to form Epo-independent colonies in response to Friend Virus infection. This defect can be rescued by exogenous expression of wild-type Gab2, but not Gab1. Moreover, we find that the Sf-Stk/Gab2 fusion protein, but not the Sf-Stk/Gab1 fusion protein, which fails to support hematopoietic transformation in this system, results in enhanced tyrosine phosphorylation of and interaction with signal transducer and activator of transcription 3 (Stat3). Our in vivo studies suggest that Stat3 is tyrosine phosphorylated and interacts with Gab2 in splenocytes following injection of sensitive mice with Friend Virus. Exogenous expression of dominant negative Stat3 in sensitive erythroblasts significantly inhibits BFU-e and CFU-e colony formation induced by Friend Virus. Furthermore, bone marrow cells from floxed-Stat3 mice fail to form Epo-independent colonies in response to Friend Virus infection following the retroviral introduction of Cre-recombinase into primary erythroblasts. In order to map the domains of Gab2 which are critical for the activation of Stat3 and the Epo-independent growth of Friend Virus infected cells, we utilized a series of Gab1/Gab2 chimeric molecules. Our data suggest that the region between Q120 and P358 of Gab2 is required for the tyrosine phosphorylation of Stat3. By scanning this region, we identified a Y194LHQ site, a potential Stat3 binding motif, in Gab2, but not in Gab1. The ability of Sf-Stk /Gab2 to recruit and activate Stat3, and support Epo-independent colony formation of primary erythroblasts in response to Friend Virus infection in vitro was abolished when Y194 motif was mutated to phenylalanine. In addition, mutation of this tyrosine in the context of wide type Gab2 abrogated its ability to rescue the defective response of Gab2−/− bone marrow to Friend Virus. In conclusion, these results suggest that a Sf-Stk/Grb2/Gab2/Stat3 signaling cascade serves as a key mediator in the Friend Virus induced transformation of erythroid cells and that the Y194LHQ motif of Gab2 is required for the activation of Stat3 during the transformation of primary erythroid cells in this system.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5245-5245
Author(s):  
Yaacov Ben-David ◽  
Tatiana Usenko ◽  
Mehran Haeri ◽  
Laura Vecchiarelli-Federico ◽  
Josef T. Prchal

Abstract PV is a myeloproliferative disease, displaying a pronounced increase in the number of erythroid cells. Despite this increase, PV patients exhibit a lower incidence of acute leukemia and in particular erythroleukemia compared to its related disorder CML. In this study we induced erythroleukemia in a mouse model of familiar polycythemia, carrying a truncated form of the human erythropoietin-receptor (Epo-R). Previous studies have shown that these mice develop a disease similar to patients with familiar polycythemia, from whom this gain-of-function receptor was isolated and introduced as transgene. We show that the progression of erythroleukemia induced by Friend virus is significantly delayed in this genetic mouse model of polycythemia. These mice exhibit a massive enrichment of Sca1+/cKit− progenitors and mature immune cells compared to normal mice. Co-transplantation experiments revealed that Sca1+/cKit− progenitors inhibit the robust tumorigenic potential of Sca1−/cKit+ erythroleukemic cells. In order to determine the mechanism of erythroleukemia inhibition, we established cell lines from Sca1+/cKit− cell populations and demonstrated a similar anti-leukemic inhibition in vivo. These cells also express cell surface markers for early B-cells (B220) and macrophages (Mac1), and are able to undergo differentiation into macrophages and dendritic cells following treatment with GM-CSF and IL4. We have shown that these cells also secrete factors that inhibit proliferation of erythroleukemic cells in culture, such as Nitric Oxide (NO). Accordingly, an NO inhibitor can suppress the inhibitory effect of the Sca-1+/cKit−/B220+/Mac1+ cells. To further demonstrate the importance of these leukemic inhibitory cells, we induced a polycythemia in normal mice through administration of Epo. Epo administration delayed the development of Friend virus-induced erythroleukemia and significantly increased the number of leukemic inhibitory Sca1+ cells. Our study also demonstrates a significant increase in the number of leukemic inhibitory cells with anti-proliferative effects in response to Epo and granulocyte macrophage-colony stimulating factor (GM-CSF) stimulation. We therefore propose that administration of Epo and GM-CSF in combination with NO agonistic drugs, as well as the identification of novel drugs that can induce the inhibitory Sca-1+/cKit−/B220+/Mac1+ cells, may lead to improved therapies for the treatment of myelodysplastic syndrome. Together, our results identify Sca-1+/cKit−/B220+/Mac1+ cells as potent cellular inhibitors of leukemia and unravel a potentially novel therapeutic approach.


Sign in / Sign up

Export Citation Format

Share Document