In 1957, Charlotte Friend described a novel retroviral disease in mice characterized by splenic enlargement, erythroleukemia, and death.1 This rapidly progressive disease, now known as Friend disease, has provided a powerful tool for the study of multistage carcinogenesis. Transformed murine erythroleukemia (MEL) cells, isolated from Friend virus–infected mice, have also provided a versatile model system for the study of erythroblast differentiation and erythropoietin (Epo)-induced signal transduction in vitro. Over the years, Friend disease has provided major insights into the molecular evolution of leukemia, the normal mechanisms of Epo receptor (EpoR) activation and, most recently, the molecular mechanisms of leukemia resistance. In this review, we summarize the molecular insights gained from the study of Friend disease. We focus on recent advances, with a special emphasis on studies of the Friend virus susceptibility locus,Fv2.2 

Following inoculation of a susceptible mouse with Friend virus complex, Friend disease evolves in 2 stages.3,4 In the first stage, the mouse develops massive splenic enlargement due to the polyclonal proliferation of splenic erythroblasts. These erythroblasts are not frankly transformed, however, and their transfer to another animal does not result in tumors.5 The primary mitogenic event underlying this polyclonal expansion is constitutive activation of the EpoR.4,6 During this early stage of disease, there is a marked expansion of erythroid progenitor cells (burst-forming units–erythroid [BFU-E] and colony-forming units–erythroid [CFU-E]).7,8 

The second stage of Friend disease is marked by the emergence of fully transformed cells in the spleen and, eventually, the blood, bone marrow, and liver.5 These transformed cells form spleen colonies in genetically anemic Sl/Sldmice, are tumorigenic in vivo, and give rise to stable cell lines in vitro.5,9,10 Mice infected with the polycythemia-inducing strain of Friend virus (see below) often die during the first stage of disease because of splenic rupture. Mice that survive the first stage, however, will develop leukemia.11 Most emerging leukemic clones either fail to express p53, or they express a mutant form of p53 that is inactive, demonstrating that loss of p53 function is a common event in disease progression.12-14 In addition, anets family transcriptional activator, Sfpi1/PU.1 (spleen focus-forming virus [SFFV] proviral integration 1), is activated in 70% to 95% of Friend virus–induced erythroleukemias.15,16 The high incidence of p53 inactivation and Sfpi1/PU.1 activation suggests that these events are critical to the evolution of erythroleukemia. Isolated MEL cells have the characteristic appearance of immature erythroblasts. These cells remain transformed and Epo-independent and demonstrate constitutive activation of the EpoR.17 Molecular events underlying the first and second stages of Friend disease have been reviewed.4,18,19 

Friend virus complex consists of 2 viruses, a replication-competent Friend helper virus (F-MuLV) and a replication-defective SFFV. There are at least 2 naturally occurring variants of SFFV, called SFFVP (polycythemia strain) and SFFVA (anemia strain). Mice infected with the viral complex of SFFVP and F-MuLV (also known as FVP) become polycythemic, while mice infected with SFFVA and F-MuLV (FVA) develop anemia as a result of volume expansion.20 In contrast to other acutely oncogenic retroviruses, SFFV does not carry a constitutively active, mutant form of a host gene. Instead, SFFV encodes a truncated form of a retroviral envelope protein, gp55, which can bind and activate the EpoR.6,21 Indeed, virally encoded gp55 can replace Epo in supporting the formation of erythroid colonies in vitro, but only in the presence of the EpoR.22 

The polycythemia- and anemia-inducing strains of Friend virus exhibit differential transforming effects in vitro.23 Both SFFVP and SFFVA induce BFU-E in methylcellulose in the absence of exogenous Epo. However, in contrast to SFFVP, SFFVA-induced bursts require Epo to undergo terminal differentiation. FVA-infected erythroblasts proliferate in vivo despite low levels of endogenous Epo.24 Withdrawal of Epo from FVA-infected erythroblasts at the CFU-E stage results in rapid commitment to undergo apoptosis.24 Thus, while FVA-infected erythroblasts can proliferate in the absence of Epo, Epo is required for their survival and terminal differentiation. Erythroblasts isolated from the spleens of FVA-infected mice (FVA cells) provide an excellent model system for the study of Epo-dependent terminal differentiation in vitro.25 

Early evidence from several investigators, including Hankins, Ruscetti, and Kabat, demonstrated that SFFV is the causative agent in the first stage of Friend disease. In fact, the gp55 oncoprotein alone was shown to induce BFU-E in vitro.26 Moreover, gp55P (from SFFVP) supports the growth of Epo-dependent cell lines in vitro, suggesting that gp55Pmight activate the cell surface EpoR directly.27 

The next clue to the function of the gp55 envelope arose from the cloning of the EpoR itself. The murine EpoR was expression-cloned from Epo-independent MEL cells expressing cell surface Epo-binding proteins.28 The EpoR polypeptide was shown to be the first member of the cytokine receptor superfamily, which includes the receptors for interleukin (IL)-2, IL-3, granulocyte-macrophage colony-stimulating factor, and several other hematopoietic cytokines.29,30 Initially, the EpoR was heterologously expressed in an IL-3–dependent, pro-B lymphocyte line, Ba/F3. Ba/F3-EpoR cells could be grown in either Epo or IL-3, demonstrating that the EpoR activates the appropriate signal transduction pathways in these cells. Next, Ba/F3-EpoR cells were infected with a retrovirus encoding gp55P. The EpoR and gp55P were shown to form a physical and functional complex in these cells, resulting in their transformation to growth factor independence. Expression of gp55 in parental Ba/F3 cells, in the absence of the EpoR, failed to transform the cells, demonstrating that both proteins are required for Epo-independent growth.6 

The nature of the molecular interaction between gp55P and the EpoR is now understood in considerable detail. Gp55Pbinds to the EpoR both at the cell surface31 and within the endoplasmic reticulum32 of Friend virus–infected erythroblasts. Binding of gp55P results in the stabilization and constitutive activation of the EpoR.32Gp55P selectively activates the murine EpoR but not the human EpoR, despite the high (92%) amino acid identity between these polypeptides.33,34 Gp55P does not activate other members of the cytokine receptor superfamily. The critical contact between gp55P and the murine EpoR occurs within the transmembrane region of these cell surface proteins,35-37and a productive interaction between the EpoR and gp55Poccurs when both proteins are expressed at the cell surface.35,37-39 The gp55P-EpoR interaction cannot be transmitted by cell-to-cell contact or by the administration of soluble gp55P to EpoR-expressing cells (A.D., unpublished observations, September 1999).

The gp55 envelope proteins of SFFVA and SFFVPdiffer in a small number of amino acid residues, primarily in the transmembrane region, and these differences appear to account for the phenotypic variation of the disease.35,36,40Gp55A does not support the growth of Epo-dependent cell lines, which raises the question of whether it functionally interacts with the EpoR. Gp55 from Rauscher leukemia virus (R-gp55; which is closely related to gp55A) physically associates with the EpoR.34 However, R-gp55 does not detectably interact with the EpoR at the cell surface.41 Part of the explanation for these differences may be that gp55A and R-gp55, when compared with gp55P, are expressed at low levels on the cell surface.42 Recently, it was shown that gp55A induces the formation of CFU-E from fetal liver cells without exogenous Epo.22 Neither gp55A nor gp55P were able to induce CFU-E formation from fetal liver cells that lacked the EpoR. Thus, the EpoR is required for gp55A-induced erythroid colony formation. Whether this reflects a requirement for a direct interaction between gp55A and the EpoR remains to be determined.

The specific interaction between gp55P and the EpoR underscores the importance of the EpoR in the evolution of erythroleukemia. The EpoR can be constitutively activated by other molecular mechanisms, also resulting in the development of erythroleukemia. For instance, the EpoR can be activated by point mutations34,43 or by erythroblast autocrine secretion of Epo.44 Indeed, a constitutively active form of the EpoR can result in induction of Friend-like disease.45 Other molecular events resulting in up-regulation of EpoR activity, such as insertional activation of EpoR expression,46,47 truncation of the EpoR carboxy terminus,48,49 or cellular loss of the negative regulatory phosphatase, Shp-1,50-53 may also contribute to the evolution of erythroleukemia.

Although the EpoR polypeptide has been molecular cloned,28 there are still questions about the structure and composition of the cell surface EpoR complex. Several studies indicate that the EpoR is a simple monomeric cytokine receptor capable of homodimerization in response to Epo binding. A soluble secreted form of the EpoR is sufficient for high-affinity Epo binding, supporting a model of a single receptor subunit.54 The crystal structure confirms the homodimeric structure of the Epo-bound, activated EpoR.55,56 Additional studies suggest a more complex membrane-bound EpoR conformation.57,58 Small synthetic peptides are capable of activating EpoR dimerization, providing a possible avenue for Epo-mimetic drug design.59,60 

Other studies suggest that the native cell surface EpoR complex may contain additional subunits or accessory proteins. First, the EpoR displays at least 2 different affinities for Epo, raising the possibility of distinct receptor classes.61,62 Second, cross-linking studies with 125I-Epo reveal multiple classes of cell surface EpoR, suggesting the presence of additional receptor subunits.31,63-65 In one study, the EpoR was cross-linked to a cell surface tyrosine kinase protein.66 In addition, the EpoR interacts with a cytoplasmic tyrosine kinase, Jak2.67 Third, forced expression of the EpoR complementary DNA in some lymphoid cell lines, such as CTLL68,69 and HT-2,70 confers Epo binding but not Epo-induced mitogenesis. These cells lack a required factor in the trans-acting environment.70 One study suggests that the Kit receptor tyrosine kinase activates the EpoR directly, thereby accounting for the synergistic activities of Epo and Kit ligand (Mgf), also known as SCF, in erythropoiesis.71 Emerging data discussed below suggest that a truncated form of the Stk receptor tyrosine kinase, encoded by the Fv2 Friend virus susceptibility locus, may also function as a subunit of the EpoR complex.2 

A number of host genes have been identified that affect the susceptibility of mice to Friend virus–induced erythroleukemia (Table1). These genes can be divided into several categories based on their proposed mechanism of resistance.72 The first group consists of genes that dominantly interfere with the infection of target cells by the retrovirus. Fv4 encodes a protein related to the MuLV Env protein, which blocks ecotropic retroviral receptors.73,Fv1 encodes a Gag-related protein, which interferes with the retroviral life cycle.74 The second group consists of genes that alter the immune response to Friend virus infection.Fv3 affects the susceptibility of mice to Friend virus–induced immunosuppression.75 TwoH2-linked loci, Rfv1 and Rfv2, and one non–H2-linked locus, Rfv3, affect the recovery from Friend virus infection.76 The third group consists of genes that affect the potential of infected target cells to proliferate and differentiate. Fv5 determines whether FVP will cause anemia or polycythemia in certain strains of mice.77Mutations in Kit and Mgf either diminish the number of available target cells or their potential to proliferate.78,79 These genes are required for normal erythropoiesis, emphasizing the importance of pre-existing erythropoietic pathways for the development of Friend disease.

Table 1.

Host genes that affect susceptibility to Friend virus–induced disease

Gene  Function
Group 1: interference with retroviral infection Fv4 Blocks retroviral cell surface receptors  
 Fv1 Interferes with retroviral life cycle  
Group 2: altered immune response Fv3 Susceptibility to Friend virus-induced immunosuppression  
 Rfv1, Rfv2 H2-linked determinants of immune response  
 Rfv3 Non–H2-linked determinant of immune response  
Group 3: regulators of erythroid cell proliferation and differentiation Fv5 Determines anemia vs polycythemia  
 Kit, Mgf Required for normal erythropoiesis 
 Fv2 Determines proliferative response to gp55 
Gene  Function
Group 1: interference with retroviral infection Fv4 Blocks retroviral cell surface receptors  
 Fv1 Interferes with retroviral life cycle  
Group 2: altered immune response Fv3 Susceptibility to Friend virus-induced immunosuppression  
 Rfv1, Rfv2 H2-linked determinants of immune response  
 Rfv3 Non–H2-linked determinant of immune response  
Group 3: regulators of erythroid cell proliferation and differentiation Fv5 Determines anemia vs polycythemia  
 Kit, Mgf Required for normal erythropoiesis 
 Fv2 Determines proliferative response to gp55 

Fv2 is another host factor that affects the proliferation of SFFV-infected erythroblasts.80 Susceptibility at theFv2 locus, which is dominant, is present in most inbred strains of mice. The exception is C57BL/6 and related strains, which are Fv2-resistant (Fv2rr). Fv2 does not interfere with retroviral entry into cells or the retroviral life cycle.81 Instead, Fv2 determines the potential of SFFV-infected erythroblasts to proliferate in response to gp55.Fv2 acts in a cell autonomous manner.82Indirect evidence suggests that Fv2 and gp55 are functionally interrelated. Fv2-mediated resistance can be circumvented by deletions within the ecotropic domain of gp55, but the same mutants are less active than wild-type gp55 inFv2-sensitive (Fv2ss) mice.83 

Recently it has been shown that Fv2 is Ron, a member of the Met subfamily of receptor tyrosine kinases.2,Ron encodes the Stk receptor tyrosine kinase, and a naturally expressed truncated form of Stk (SF-Stk) confers susceptibility to Friend virus–induced erythroleukemia. The evidence can be summarized as follows. First, the expression of SF-Stk correlates with susceptibility to Friend disease. SF-Stk is the major expressed form of Stk in Fv2ss erythroid cells; however, SF-Stk is expressed at very low levels or is absent inFv2rr erythroid cells. The 5′ end of the SF-Stk transcript has been mapped to intron 10 ofRon.84 The presumptive basis for the difference in expression is a 3-nucleotide deletion in C57BL/6 DNA, which mutates a GATA binding site and causes a 6-fold difference in promoter activity. Second, targeted disruption of Stk inFv2ss mice causes resistance to Friend disease. Third, enforced expression of SF-Stk in Fv2rrmice confers susceptibility to Friend disease. These data strongly suggest that SF-Stk confers susceptibility to gp55-mediated erythroblastosis and that C57BL/6 mice are resistant because they fail to express the protein.

The molecular mechanism by which SF-Stk confers susceptibility to Friend disease is not known. Possibly, full-length Stk would have the same effect as SF-Stk if expressed at an equivalent level. Alternatively, there may a unique requirement for SF-Stk. The predicted protein encoded by the SF-Stk transcripts lacks the extracellular domain of Stk but retains the transmembrane and kinase domains.84 By analogy to other receptor tyrosine kinases, loss of the extracellular domain may contribute to constitutive activation.85 SF-Stk is closely related to the avian oncogene v-sea, which causes erythroblastosis and anemia in chickens.86 The v-sea oncogene is a transmembrane protein, which has an extracellular domain related to an avian retroviral envelope and an intracellular domain derived from c-sea. One possible mechanism is that gp55 and SF-Stk together form the functional equivalent of v-sea. Crosstalk could occur between gp55, SF-Stk, and components of the EpoR complex (eg, Jak2), resulting in activation of downstream signaling cascades. In support of this model, SF-Stk and the EpoR have been shown to physically interact in COS cells.87 Based on differences in molecular weight, Stk does not appear to be one of the proteins previously identified by 125I-Epo cross-linking to the EpoR.31,63-65 A second possibility is that SF-Stk functions independently of the gp55-EpoR complex to enhance erythroid cell survival or proliferation.

The lack of an erythroid defect in (Fv2rr) C57BL/6 mice suggests that SF-Stk does not have a role in normal erythropoiesis. Still, the role of full-length Stk remains to be determined. Stk is the receptor for macrophage-stimulating protein (also known as hepatocyte growth factor–like protein or Hgfl).88 Targeted disruption of the first exon of Stk in mice causes increased nitric oxide production from peritoneal macrophages in response to interferon-γ.89Under normal conditions, these mice are viable and the mutation has no effect on erythropoiesis. In contrast, deletion of a large part of the Stk gene is associated with early embryonic lethality due to a failure of implantation.90 One explanation for these findings may be that the exon 1 mutation of Stk is a hypomorphic allele rather than a null allele of Stk. Studies in mice that are chimeric for Stk-deficient cells should resolve whether Stk has a role in normal erythropoiesis.

The Met subfamily of receptor tyrosine kinases includes Met, Ron, Stk (the murine homolog of Ron), and v-sea. These receptors have a distinctive 2-tyrosine docking site in the C-terminal tail that mediates interactions with multiple SH2-containing signal transducers.91 Ligand binding elicits a complex biologic response that includes cell growth, dissociation, motility, and polarization.92 Recent evidence suggests that the Met subfamily may function as second subunits or coreceptors for the cytokine receptor family. Stk has been shown to colocalize with and activate the IL-3 receptor common β-chain, another member of the cytokine receptor family.93 Hgf, the ligand for Met, has a synergistic effect with Epo on Stat5 activation and erythroid burst formation, suggesting that Met can productively interact with the EpoR.94 Finally, it is intriguing that another member of the Met family, v-sea, causes erythroblastosis and anemia in chickens.86 Studies on potential interactions between v-sea and the avian EpoR await cloning of the avian EpoR.

Based on the likely physical and functional interaction between the EpoR and Stk,87 several questions emerge. For instance, it will be interesting to determine (1) whether activation of the EpoR results in activation of Stk kinase activity or phosphorylation of the multifunctional docking site; (2) whether activation of Stk by Hgfl results in EpoR phosphorylation or activation of Jak2; (3) whether EpoR and Stk can be cross-linked at the cell surface or whether previously identified Epo/EpoR cross-linked complexes62 contain Stk; and (4) whether specific activation or phosphorylation events correlate with the progression of Friend disease.

The molecular events underlying the first and second stages of Friend disease are now understood in considerable detail (Figure1). In the first stage of Friend disease, constitutive activation of the EpoR by gp55 causes uncontrolled, polyclonal proliferation of infected erythroblasts (Figure 1A). Susceptibility to gp55-induced erythroblastosis depends on expression of SF-Stk. Recent studies suggest that the mechanism may involve a direct interaction between the EpoR and SF-Stk.87 In the second stage of Friend disease, activation of Sfpi1/PU.1 and loss of p53 contribute to erythroleukemic transformation (Figure 1B and Table2). The transformation-specific targets of Sfpi1/PU.1 are not known, although one potentially relevant target is Fli1.95 Another potential target is SF-Stk. SF-Stk expression is down-regulated during terminal differentiation of FVA-infected, primary erythroblasts (P.N., unpublished observations, December 1998). Potentially, Sfpi1/PU.1 could activate SF-Stk through Ets sites in the SF-Stk promoter.2Constitutive SF-Stk expression, in turn, could contribute to the immortalization of infected erythroblasts. Consistent with this idea, SF-Stk is highly expressed in MEL cells but not most other hematopoietic cell lines. Further, enforced expression of full-length Stk causes ligand-dependent apoptosis of MEL cells but not Ba/F3 cells, indicating that Stk signaling has a direct effect on MEL cell survival.96 

Fig. 1.

A speculative model of multistage Friend disease resulting from an interaction of the EpoR and SF-Stk.

(A) In the first stage of the disease, an SFFV provirus integrates into the host genome and encodes the oncogenic envelope protein, gp55. Then, gp55 forms a high-molecular-weight complex with the EpoR and perhaps SF-Stk, both at the cell surface and within an intracellular compartment. This binding interaction leads to the constitutive activation of the EpoR. SF-Stk may contribute to this mechanism in one or more ways. For instance, SF-Stk may increase the affinity of EpoR-gp55 interaction, increase the expression of cell surface EpoR, act as a coreceptor that activates additional downstream signaling events, or activate unique downstream signaling events that are required for erythroblast transformation. (B) In the second stage of the disease, proviral integrations cause up-regulation of Sfpi1/PU.1 expression and inactivation of wild-type p53 as well as other events (not shown).101 These mutations confer a selective advantage. Although this remains speculative, Sfpi1/PU.1 may activate SF-Stk as well as other target genes, resulting in progression toward erythroleukemia.

Fig. 1.

A speculative model of multistage Friend disease resulting from an interaction of the EpoR and SF-Stk.

(A) In the first stage of the disease, an SFFV provirus integrates into the host genome and encodes the oncogenic envelope protein, gp55. Then, gp55 forms a high-molecular-weight complex with the EpoR and perhaps SF-Stk, both at the cell surface and within an intracellular compartment. This binding interaction leads to the constitutive activation of the EpoR. SF-Stk may contribute to this mechanism in one or more ways. For instance, SF-Stk may increase the affinity of EpoR-gp55 interaction, increase the expression of cell surface EpoR, act as a coreceptor that activates additional downstream signaling events, or activate unique downstream signaling events that are required for erythroblast transformation. (B) In the second stage of the disease, proviral integrations cause up-regulation of Sfpi1/PU.1 expression and inactivation of wild-type p53 as well as other events (not shown).101 These mutations confer a selective advantage. Although this remains speculative, Sfpi1/PU.1 may activate SF-Stk as well as other target genes, resulting in progression toward erythroleukemia.

Close modal
Table 2.

Molecular events associated with progression of Friend virus-induced erythroleukemia

GeneOrigin of cell line or tumorGenetic changeFrequencyReferences
PU.1(Sfpi1FVP/FVA Proviral insertion, ↑Pu.1 70%-95% 15, 16  
Fli1 F-MuLV Proviral insertion, ↑Fli-1 75%-90% 97, 98  
Fli2(Nfe2FVP, F-MuLV Proviral insertion, ↓Nfe2 10% 101  
Epo F-MuLV Genomic rearrangement, ↑Epo 80% 100  
EpoR FVP Proviral insertion, ↑EpoR Unknown 46, 47  
p53(Trp53FVP/FVA, F-MuLV 1. Proviral insertion, ↓p53 ∼ 100% 12-14  
  2. Genomic rearrangement, ↓p53, truncation of p53   
  3. Point mutation, loss of normal p53 function   
GeneOrigin of cell line or tumorGenetic changeFrequencyReferences
PU.1(Sfpi1FVP/FVA Proviral insertion, ↑Pu.1 70%-95% 15, 16  
Fli1 F-MuLV Proviral insertion, ↑Fli-1 75%-90% 97, 98  
Fli2(Nfe2FVP, F-MuLV Proviral insertion, ↓Nfe2 10% 101  
Epo F-MuLV Genomic rearrangement, ↑Epo 80% 100  
EpoR FVP Proviral insertion, ↑EpoR Unknown 46, 47  
p53(Trp53FVP/FVA, F-MuLV 1. Proviral insertion, ↓p53 ∼ 100% 12-14  
  2. Genomic rearrangement, ↓p53, truncation of p53   
  3. Point mutation, loss of normal p53 function   

It is interesting to contrast the progression of FVP-induced disease with that of F-MuLV, which is gp55-independent. F-MuLV–induced erythroleukemia has a longer latency (6 weeks) than FVP-induced disease (1-2 weeks), is oligoclonal rather than polyclonal and is associated with the activation of Fli1 rather than Sfpi1/PU.1 expression.97-99 Most MEL cell lines derived from Fli1-expressing primary erythroid tumors are Epo-dependent. However, with additional passages in vivo Epo-independent MEL lines are obtained, which show a high frequency of genomic rearrangements inEpo and autonomous Epo expression.100Thus, whether it occurs early in the course of disease (FVP) or late (F-MuLV), deregulation of EpoR signaling appears to be a common step in the progression of Friend virus–induced erythroleukemia.

In summary, Friend virus–induced erythroleukemia has provided a model system for the molecular understanding of the multistage nature of leukemogenesis. Critical elements of the disease involve the interaction of host gene products (EpoR, Sfpi1/PU.1, and SF-Stk) and retroviral gene products (gp55P or gp55A). The recent demonstration of a specific role of the cell surface tyrosine kinase SF-Stk in the process of leukemogenesis underscores the importance of multiple signal transduction pathways in normal hematopoiesis and leukemogenesis.

Stk is a member of a large family of cell surface tyrosine kinase receptors, while the EpoR is a member of a large family of cytokine receptors. Accordingly, it will be interesting to determine whether other transmembrane tyrosine kinases and cytokine receptors interact at the cell surface of hematopoietic cells. These tyrosine kinases may have a more general role as modifiers (coreceptors) of cytokine receptor signaling. In this regard, it is interesting thatKit encodes a receptor tyrosine kinase that is essential for normal erythropoiesis. Mgf and Epo are potent comitogens for early erythroid progenitor cells, and this effect may be due to direct interaction between Kit and the EpoR.71 

Finally, identification of SF-Stk as the product of the Fv2gene provides general insights into the mechanism of cancer resistance in mice and perhaps in humans. Friend disease develops when the proliferation of infected erythroblasts outpaces the ability of the host immune system to suppress the infection.76 Mice that are deficient in SF-Stk, such as the C57BL/6 strain, have lost little or none of their normal erythropoietic response; however, gp55-mediated erythroid proliferation is slowed. As a result, they gain resistance to Friend virus–induced erythroleukemia.

We thank James Ihle, Dwayne Barber, and Maurice Bondurant for helpful discussions and advice on the manuscript.

Supported by National Institutes of Health (NIH) grant RO1 CA84214 (P.A.N.), NIH Cancer Center Support Grant P30 CA21765, and the American Lebanese Syrian Associated Charities.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Friend
C
Cell-free transmission in adult swiss mice of a disease having the character of a leukemia.
J Exp Med.
105
1956
307
318
2
Persons
DA
Paulson
RF
Loyd
MR
et al
Fv2 is a truncated form of the Stk/RON receptor tyrosine kinase.
Nat Genet.
23
1999
159
165
3
Ben-David
Y
Bernstein
A
Friend virus-induced erythroleukemia and the multistage nature of cancer.
Cell.
66
1991
831
834
4
D'Andrea
AD
The interaction of the erythropoietin receptor and gp55. Oncogenes in the development of leukaemia.
Cancer Surv.
15
1992
19
35
5
Wendling
F
Moreau-Gachelin
F
Tambourin
P
Emergence of tumorigenic cells during the course of Friend virus leukemias.
Proc Natl Acad Sci U S A.
78
1981
3614
3618
6
Li
J-P
D'Andrea
AD
Lodish
HF
Baltimore
D
Activation of cell growth by binding of Friend spleen focus-forming virus gp55 glycoprotein to the erythropoietin receptor.
Nature.
343
1990
762
764
7
Liao
SK
Axelrad
AA
Erythropoietin-independent erythroid colony formation in vitro by hemopoietic cells of mice infected with friend virus.
Int J Cancer.
15
1975
467
482
8
Peschle
C
Migliaccio
G
Lettieri
F
et al
Kinetics of erythroid precursors in mice infected with the anemic or the polycythemic strain of Friend leukemia virus.
Proc Natl Acad Sci U S A.
77
1980
2054
2058
9
Mager
D
Mak
TW
Bernstein
A
Friend leukaemia virus-transformed cells, unlike normal stem cells, form spleen colonies in Sl/Sld mice.
Nature.
288
1980
592
594
10
Mager
DL
Mak
TW
Bernstein
A
Quantitative colony method for tumorigenic cells transformed by two distinct strains of Friend leukemia virus.
Proc Natl Acad Sci U S A.
78
1981
1703
1707
11
Mirand
EA
Hoffman
JG
Grace
JT
Jr
Trudel
PJ
Modification of the Friend virus disease by splenectomy.
Proc Soc Exp Biol Med.
107
1961
824
828
12
Mowat
M
Cheng
A
Kimura
N
Bernstein
A
Benchimol
S
Rearrangements of the cellular p53 gene in erythroleukaemic cells transformed by Friend virus.
Nature.
314
1985
633
636
13
Ben-David
Y
Prideaux
VR
Chow
V
Benchimol
S
Bernstein
A
Inactivation of the p53 oncogene by internal deletion or retroviral integration in erythroleukemic cell lines induced by Friend leukemia virus.
Oncogene.
3
1988
179
185
14
Munroe
DG
Peacock
JW
Benchimol
S
Inactivation of the cellular p53 gene is a common feature of Friend virus-induced erythroleukemia: relationship of inactivation to dominant transforming alleles.
Mol Cell Biol.
10
1990
3307
3313
15
Moreau-Gachelin
F
Tavitian
A
Tambourin
P
Spi-1 is a putative oncogene in virally induced murine erythroleukemias.
Nature.
331
1988
277
280
16
Paul
R
Schuetze
S
Kozak
SL
Kabat
D
A common site for immortalizing proviral integrations in Friend erythroleukemia: molecular cloning and characterization.
J Virol.
63
1989
4958
4961
17
Yamamura
Y
Senda
H
Kageyama
Y
Matsuzaki
T
Noda
M
Ikawa
Y
Erythropoietin and Friend virus gp55 activate different JAK/STAT pathways through the erythropoietin receptor in erythroid cells.
Mol Cell Biol.
18
1998
1172
1180
18
Kabat
D
Molecular biology of Friend viral erythroleukemia.
Curr Top Microbiol Immunol.
148
1989
1
42
19
Ruscetti
S
Erythroleukaemia induction by the Friend spleen focus-forming virus.
Baillieres Clin Haematol.
8
1995
225
247
20
Tambourin
PE
Gallien-Lartigue
O
Wendling
F
Huaulme
D
Erythrocyte production in mice infected by the polycythaemia-inducing Friend virus or by the anaemia-inducing Friend virus.
Br J Haematol.
24
1973
511
524
21
Hoatlin
ME
Kozak
SL
Lilly
F
Chakraborti
A
Kozak
CA
Kabat
D
Activation of erythropoietin receptors by Friend viral gp55 and by erythropoietin and down-modulation by the murine Fv-2r resistance gene.
Proc Natl Acad Sci U S A.
87
1990
9985
9989
22
Constantinescu
SN
Wu
H
Liu
X
Beyer
W
Fallon
A
Lodish
HF
The anemic Friend virus gp55 envelope protein induces erythroid differentiation in fetal liver colony-forming units-erythroid.
Blood.
91
1998
1163
1172
23
Hankins
WD
Troxler
D
Polycythemia and anemia-inducing erythroleukemia viruses exhibit differential erythroid transforming effects in vitro.
Cell.
22
1980
693
699
24
Koury
MJ
Bondurant
MC
Erythropoietin retards DNA breakdown and prevents programmed death in erythroid progenitor cells.
Science.
248
1990
378
381
25
Koury
M
Sawyer
S
Bondurant
M
Splenic erythroblasts in anemia-inducing Friend disease: a source of cells for studies of erythropoietin-mediated differentiation.
J Cell Physiol.
121
1984
526
532
26
Wolff
L
Ruscetti
S
The spleen focus-forming virus (SFFV) envelope gene, when introduced into mice in the absence of other SFFV genes, induces acute erythroleukemia.
J Virol.
62
1988
2158
2163
27
Ruscetti
SK
Janesch
NJ
Chakraborti
A
Sawyer
ST
Hankins
WD
Friend spleen focus-forming virus induces factor independence in an erythropoietin-dependent erythroleukemia cell line.
J Virol.
63
1990
1057
1062
28
D'Andrea
AD
Lodish
HF
Wong
GG
Expression cloning of the murine erythropoietin receptor.
Cell.
57
1989
277
285
29
D'Andrea
AD
Fasman
GD
Lodish
HF
Erythropoietin receptor and interleukin receptor beta chain: a new receptor family.
Cell.
58
1989
1023
1024
30
Bazan
JF
A novel family of growth factor receptors: a common binding domain in the growth hormone, prolactin, erythropoietin and IL-6 receptors, and the p75 IL-2 receptor β-chain.
Biochem Biophys Res Commun.
164
1989
788
795
31
Casadevall
N
Lacombe
C
Muller
O
Gisselbrecht
S
Mayeux
P
Multimeric structure of the membrane erythropoietin receptor of murine erythroleukemia cells.
J Biol Chem.
266
1991
16015
16020
32
Yoshimura
A
D'Andrea
AD
Lodish
HF
The Friend spleen focus-forming virus glycoprotein interacts with the erythropoietin receptor in an intracellular compartment and affects receptor metabolism.
Proc Natl Acad Sci U S A.
87
1990
4139
4143
33
Jones
SS
D'Andrea
AD
Haines
LL
Wong
GG
Human erythropoietin receptor: cloning expression and biologic characterization.
Blood.
76
1990
31
35
34
Showers
MO
DeMartino
JC
Saito
Y
D'Andrea
AD
Fusion of the erythropoietin receptor and the Friend spleen focus-forming virus gp55 glycoprotein transforms a factor-dependent hematopoietic cell line.
Mol Cell Biol.
13
1993
739
748
35
Chung
S-W
Wolff
L
Ruscetti
SK
Transmembrane domain of the envelope gene of a polycythemia-inducing retrovirus determines erythropoietin-independent growth.
Proc Natl Acad Sci U S A.
86
1989
7957
7960
36
Constantinescu
SN
Liu
X
Beyer
W
et al
Activation of the erythropoietin receptor by the gp55-P viral envelope protein is determined by a single amino acid in its transmembrane domain.
EMBO J.
18
1999
3334
3347
37
Zon
LI
Moreau
JF
Koo
JW
Mathey-Prevot
B
D'Andrea
AD
The erythropoietin receptor transmembrane region is necessary for activation by the Friend spleen focus-forming virus gp55 glycoprotein.
Mol Cell Biol.
12
1992
2949
2957
38
Ferro
FE
Kozak
SL
Hoatlin
ME
Kabat
D
Cell surface site for mitogenic interaction of erythropoietin receptors with the membrane glycoprotein encoded by Friend erythroleukemia virus.
J Biol Chem.
268
1993
5741
5747
39
Li
JP
Hu
HO
Niu
QT
Fang
C
Cell surface activation of the erythropoietin receptor by Friend spleen focus-forming virus gp55.
J Virol.
69
1995
1714
1719
40
Fang
C
Choi
E
Nie
L
Li
JP
Role of the transmembrane sequence of spleen focus-forming virus gp55 in erythroleukemogenesis.
Virology.
252
1998
46
53
41
Tarr
K
Watowich
SS
Longmore
GD
Cell surface organization of the erythropoietin receptor complex differs depending on its mode of activation.
J Biol Chem.
272
1997
9099
9107
42
Ruscetti
SK
Feild
JA
Scolnick
EM
Polycythaemia- and anaemia-inducing strains of spleen focus-forming virus differ in post-translational processing of envelope-related glycoproteins.
Nature.
294
1981
663
665
43
Yoshimura
A
Longmore
G
Lodish
HF
Point mutation in the exoplasmic domain of the erythropoietin receptor resulting in hormone-independent activation and tumorigenicity.
Nature.
348
1990
647
649
44
McDonald
J
Beru
N
Goldwasser
E
Rearrangement and expression of erythropoietin genes in transformed mouse cells.
Mol Cell Biol.
7
1987
365
370
45
Longmore
GD
Lodish
HF
An activating mutation in the murine erythropoietin receptor induces erythroleukemia in mice: a cytokine receptor superfamily oncogene.
Cell.
67
1991
1089
1102
46
Hino
M
Tojo
A
Misawa
Y
Morii
H
Takaku
F
Shibuya
M
Unregulated expression of the erythropoietin receptor caused by insertion of spleen focus-forming virus long terminal repeat in a murine erythroleukemia cell line.
Mol Cell Biol.
11
1991
5527
5533
47
Lacombe
C
Chretien
S
Lemarchandel
U
et al
Spleen focus-forming virus long terminal repeat insertional activation of the murine erythropoietin receptor gene in the T3Cl-2 Friend leukemia cell line.
J Biol Chem.
266
1991
6952
6956
48
D'Andrea
AD
Yoshimura
A
Youssoufian
H
Zon
LI
Koo
J-W
Lodish
HF
The cytoplasmic region of the erythropoietin receptor contains nonoverlapping positive and negative growth regulatory domains.
Mol Cell Biol.
11
1991
1980
1987
49
Ward
JC
Harris
KW
Penny
LA
Forget
BG
Kitamura
T
Winkelmann
JC
A structurally abnormal erythropoietin receptor gene in a human erythroleukemia cell line.
Exp Hematol.
20
1992
371
373
50
Klingmuller
U
Lorenz
U
Cantley
L
Neel
B
Lodish
H
Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals.
Cell.
80
1995
729
738
51
Shultz
LD
Schweitzer
PA
Rajan
TV
Yi
T
Ihle
JN
Matthews
RJ
Mutations at the murine motheaten locus are within the hematopoietic cell protein-tyrosine phosphatase (Hcph) gene.
Cell.
73
1993
1445
1454
52
Tsui
HW
Siminovitch
KA
de Souza
L
Tsui
FW
Motheaten and viable motheaten mice have mutations in the haematopoietic cell phosphatase gene.
Nat Genet.
4
1993
124
129
53
Van Zant
G
Shultz
LD
Hematopoietic abnormalities of the immunodeficient mouse mutant, viable motheaten (mev).
Exp Hematol.
17
1989
81
87
54
Schimmenti
LA
Blechert
G
Harris
KW
Winkelmann
JC
Localization of an essential ligand binding determinant of the human erythropoietin receptor to a domain N-terminal to the WSXWS motif: implications for soluble receptor function.
Exp Hematol.
23
1995
1341
1346
55
Livnah
O
Johnson
DL
Stura
EA
et al
An antagonist peptide-EPO receptor complex suggests that receptor dimerization is not sufficent for activation.
Nat Struct Biol.
5
1998
993
1004
56
Livnah
O
Stura
EA
Middleton
SA
Johnson
DL
Jolliffe
LK
Wilson
IA
Crystallographic evidence for preformed dimers of erythropoietin receptor before ligand activation.
Science.
283
1999
987
990
57
Cheetham
JC
Smith
DM
Aoki
KH
et al
NMR structure of human erythropoietin and a comparison with its receptor bound conformation.
Nat Struct Biol.
5
1998
861
866
58
Syed
RS
Reid
SW
Li
C
et al
Efficiency of signalling through cytokine receptors depends critically on receptor orientation.
Nature.
395
1998
511
516
59
Livnah
O
Stura
E
Johnson
DL
et al
Functional mimicry of a protein hormone by a peptide agonist: the EPO receptor complex at 2.8 A.
Science.
273
1996
464
471
60
Qureshi
SA
Kim
RM
Konteatis
Z
et al
Mimicry of erythropoietin by a nonpeptide molecule.
Proc Natl Acad Sci U S A.
96
1999
12156
12161
61
Dong
YJ
Goldwasser
E
Evidence for an accessory component that increases the affinity of the erythropoietin receptor.
Exp Hematol.
21
1993
483
486
62
Sawyer
ST
Krantz
SB
Luna
J
Identification of the receptor for erythropoietin by cross-linking to Friend virus-infected erythroid cells.
Proc Natl Acad Sci U S A.
84
1987
3690
3694
63
Krystal
G
Humphries
K
Damen
J
A second erythropoietin receptor subunit.
Blood.
85
1995
2641
64
Mayeux
P
Lacombe
C
Casadevall
N
Chretien
S
Dusanter
I
Gisselbrecht
S
Structure of the murine erythropoietin receptor complex. Characterization of the erythropoietin cross-linked proteins.
J Biol Chem.
266
1991
23380
23385
65
Miura
O
Ihle
JN
Subunit structure of the erythropoietin receptor analyzed by 125I-EPO cross-linking in cells expressing wild-type or mutant receptors.
Blood.
81
1993
1739
1744
66
Yoshimura
A
Lodish
HF
In vitro phosphorylation of the erythropoietin receptor and an associated protein, pp130.
Mol Cell Biol.
12
1992
706
715
67
Witthuhn
BA
Quelle
FW
Silvennoinen
O
et al
JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin.
Cell.
74
1993
227
236
68
Barber
DL
D'Andrea
AD
Erythropoietin and interleukin-2 activate distinct JAK kinase family members.
Mol Cell Biol.
14
1994
6506
6514
69
Yamamura
Y
Kageyama
U
Matuzaki
T
Noda
M
Ikawa
Y
Distinct downstream signaling mechanism between erythropoietin receptor and interleukin-2 receptor.
EMBO J.
11
1992
4909
4915
70
Gaffen
SL
Lai
SY
Longmore
GD
Liu
KD
Goldsmith
MA
Genetic evidence for an additional factor required for erythropoietin-induced signal transduction.
Blood.
94
1999
74
86
71
Wu
H
Klingmuller
U
Besmer
P
Lodish
HF
Interaction of the erythropoietin and stem-cell-factor receptors.
Nature.
377
1995
242
246
72
Axelrad
A
Genetic and cellular basis of susceptibility or resistance to Friend leukemia virus infection in mice.
Can Cancer Conf.
8
1969
313
343
73
Ikeda
H
Laigret
F
Martin
MA
Repaske
R
Characterization of a molecularly cloned retroviral sequence associated with Fv-4 resistance.
J Virol.
55
1985
768
777
74
Best
S
Le Tissier
P
Towers
G
Stoye
JP
Positional cloning of the mouse retrovirus restriction gene Fvl.
Nature.
382
1996
826
829
75
Kumar
V
Resnick
P
Eastcott
JW
Bennett
M
Mechanism of genetic resistance to Friend virus leukemia in mice. V. Relevance of Fv-3 gene in the regulation of in vivo immunosuppression.
J Natl Cancer Inst.
61
1978
1117
1123
76
Hasenkrug
K
Chesebro
B
Immunity to retroviral infection: the Friend virus model.
Proc Natl Acad Sci U S A.
94
1997
7811
7816
77
Shibuya
T
Mak
TW
A host gene controlling early anaemia or polycythaemia induced by Friend erythroleukaemia virus.
Nature.
296
1982
577
579
78
Steeves
RA
Bennett
M
Mirand
EA
Cudkowicz
G
Genetic control by the W locus of susceptibility to (Friend) spleen focus-forming virus.
Nature.
218
1968
372
374
79
Bennett
M
Steeves
RA
Cudkowicz
G
Mirand
EA
Russell
LB
Mutant Sl alleles of mice affect susceptibility to Friend spleen focus-forming virus.
Science.
162
1968
564
565
80
Lilly
F
Fv-2: identification and location of a second gene governing the spleen focus response to Friend leukemia virus in mice.
J Natl Cancer Inst.
45
1970
163
169
81
Bondurant
M
Koury
MJ
Krantz
SB
The Fv-2 gene controls induction of erythroid burst formation by Friend virus infection in vitro: studies of growth regulators and viral replication.
J Gen Virol.
66
1985
83
96
82
Behringer
RR
Dewey
MJ
Cellular site and mode of Fv-2 gene action.
Cell.
40
1985
441
447
83
Hoatlin
ME
Ferro
FE
Jr
Geib
RW
Fox
MT
Kozak
SL
Kabat
D
Deletions in one domain of the Friend virus-encoded membrane glycoprotein overcome host range restrictions of erythroleukemia.
J Virol.
69
1995
856
863
84
Iwama
A
Okano
K
Sudo
T
Matsuda
Y
Suda
T
Molecular cloning of a novel receptor tyrosine kinase gene, STK, derived from enriched hematopoietic stem cells.
Blood.
83
1994
3160
3169
85
Hayman
MJ
Erb-B: growth factor receptor turned oncogene.
Trends Genet.
2
1986
260
262
86
Smith
DR
Vogt
PK
Hayman
MJ
The v-sea oncogene of avian erythroblastosis retrovirus S13: another member of the protein-tyrosine kinase gene family.
Proc Natl Acad Sci U S A.
86
1989
5291
5295
87
Herley
MT
D'Andrea
AD
Ney
PA
Physical and functional interactions between the erythropoietin receptor and a truncated form of the STK/RON receptor tyrosine kinase [abstract].
Blood.
94(suppl 1)
1999
652a
88
Gaudino
G
Follenzi
A
Naldini
L
et al
RON is a hetrodimeric tyrosine kinase receptor activated by the HGF homologue MSP.
EMBO J.
13
1994
3524
3532
89
Correll
PH
Iwama
A
Tondat
S
Mayrhofer
G
Suda
T
Bernstein
A
Deregulated inflammatory response in mice lacking the STK/RON receptor tyrosine kinase.
Genes Funct.
1
1997
69
83
90
Muraoka
RS
Sun
WY
Colbert
MC
et al
The Ron/STK receptor tyrosine kinase is essential for peri-implantation development in the mouse.
J Clin Invest.
103
1999
1277
1285
91
Ponzetto
C
Bardelli
A
Zhen
Z
et al
A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family.
Cell.
77
1994
261
271
92
Comoglio
PM
Boccaccio
C
The HGF receptor family: unconventional signal transducers for invasive cell growth.
Genes Cells.
1
1996
347
354
93
Mera
A
Suga
M
Ando
M
Suda
T
Yamaguchi
N
Induction of cell shape changes through activation of the IL-3 common β-chain receptor by the RON receptor-type tyrosine kinase.
J Biol Chem.
274
1999
15766
15774
94
Iguchi
T
Sogo
S
Hisha
H
et al
HGF activates signal transduction from EPO receptor on human cord blood CD34+/CD45+ cells.
Stem Cells.
17
1999
82
91
95
Starck
J
Doubeikovski
A
Sarrazin
S
et al
Spi-1/PU.1 is a positive regulator of the Fli-1 gene involved in inhibition of erythroid differentiation in friend erythroleukemic cell lines.
Mol Cell Biol.
19
1999
121
135
96
Iwama
A
Yamaguchi
N
Suda
T
STK/RON receptor tyrosine kinase mediates both apoptotic and growth signals via the multifunctional docking site conserved among the HGF receptor family.
EMBO J.
15
1996
5866
5875
97
Howard
JC
Yousefi
S
Cheong
G
Bernstein
A
Ben-David
Y
Temporal order and functional analysis of mutations within the Fli-1 and p53 genes during the erythroleukemias induced by F-MuLV.
Oncogene.
8
1993
2721
2729
98
Ben-David
Y
Giddens
EB
Bernstein
A
Identification and mapping of a common proviral integration site Fli-1 in erythroleukemia cells induced by Friend murine leukemia virus.
Proc Natl Acad Sci U S A.
87
1990
1332
1336
99
Sels
FT
Langer
S
Schulz
AS
Silver
J
Sitbon
M
Friedrich
RW
Friend murine leukaemia virus is integrated at a common site in most primary spleen tumours of erythroleukaemic animals.
Oncogene.
7
1992
643
652
100
Howard
JC
Berger
L
Bani
MR
Hawley
RG
Ben David
Y
Activation of the erythropoietin gene in the majority of F-MuLV-induced erythroleukemias results in growth factor independence and enhanced tumorigenicity.
Oncogene.
12
1996
1405
1415
101
Lu
S
Rowan
S
Bani
M
Ben-David
Y
Retroviral integration within the Fli-2 locus results in inactivation of the erythroid transcription factor NF-E2 in Friend erythroleukemias: evidence that NF-E2 is essential for globin expression.
Proc Natl Acad Sci U S A.
91
1994
8398
8402

Author notes

Alan D. D'Andrea, Dana-Farber Cancer Institute, Department of Pediatric Oncology, Harvard Medical School, 44 Binney St, Boston, MA 02115; e-mail:alan_dandrea@dfci.harvard.edu.

Sign in via your Institution