<<

Vet. Res. 38 (2007) 211–228 211 c INRA, EDP Sciences, 2007 DOI: 10.1051/vetres:2006060 Review article

Jaagsiekte Sheep (JSRV): from to lung cancer in sheep

Caroline La*,NicolasGa, Vincent Ca,b, Timothy Ga, Jean-François Ma,b, Fabienne Aa

a Université de Lyon 1, INRA, UMR754, École Nationale Vétérinaire de Lyon, IFR 128, F-69007, Lyon, France b Centre de référence des maladies orphelines pulmonaires, Hôpital Louis Pradel, Hospices Civils de Lyon, Lyon, France

(Received 11 September 2006; accepted 23 November 2006)

Abstract – Jaagsiekte Sheep Retrovirus (JSRV) is a infecting sheep. This virus is re- sponsible for a pulmonary adenocarcinoma, by transformation of epithelial cells from the bronchioli and alveoli. This cancer is similar to human bronchioloalveolar cancer (BAC), a specific form of human lung cancer for which a viral aetiology has not yet been identified. JSRV interacts with target cells through the membrane receptor Hyal2. The JSRV is simple and contains no recognised . It is now well established that the protein is oncogenic by itself, via the cytoplasmic domain of the transmembrane and some domains of the surface glycoprotein. Activation of the PI3K/Akt and MAPK pathways participates in the envelope- induced transformation. Tumour development is associated with telomerase activation. This review will focus on the induction of cancer by JSRV.

JSRV / ovine pulmonary adenocarcinoma / bronchioloalveolar cancer / type II pneumocytes / lung

Table of contents 1. Introduction ...... 212 2. OPA: a virus-induced cancer...... 212 2.1. Natural history ...... 212 2.2. JSRV: a member of the Betaretrovirus ...... 212 2.3. Virus and genomic organisation ...... 213 2.4. JSRVtropism...... 215 3. The JSRVinduced disease...... 216 3.1. Clinical presentation and histopathology ...... 216 3.2. Mechanisms of oncogenesis ...... 217 4. JSRVand human lung cancer...... 222 5. Conclusion...... 224

* Corresponding author: [email protected]

Article available at http://www.edpsciences.org/vetres or http://dx.doi.org/10.1051/vetres:2006060 212 C. Leroux et al.

1. INTRODUCTION The disease can also be efficiently trans- mitted to goats by experimental inocula- Ovine pulmonary adenocarcinoma tion [77, 81]. JSRV has been definitively (OPA) is a contagious tumour originat- demonstrated as the aetiological agent of ing in the distal lung after by OPA by experimental inoculation of par- Jaagsiekte Sheep Retrovirus (JSRV). The ticles produced from a JSRV-molecular disease was initially described in 1915 in clone [57]. South Africa and is present worldwide. OPA is present on all continents. Its ffi JSRV belongs to the Retroviridae family incidence is di cult to evaluate in the ab- and the Betaretrovirus genus. OPA is sence of an appropriate screening tool. The similar to a peculiar form of human virus is transmitted between by cancer, bronchioloalveolar cancer (BAC), close contact, mainly through aerosolized with which it shares clinical, radiologi- particles. The breeding conditions are of cal and histopathological features [51]. major importance for the dissemination of Although the molecular mechanisms of the virus. The incubation period in nat- JSRV induced- tumorigenesis are still only urally infected animals ranges between 2 partially understood, the significant recent and 4 years [78], but the cancer may be efforts in the field of OPA research rein- diagnosed as early as a few months af- force the relevance of this model for the ter birth. The incubation period may vary study of both the virological and cancerous according to the type of infection (experi- aspects of lung adenocarcinoma. mental versus spontaneous infection), and the age of the animals [78]. Interestingly, injection of tumoural tissues into newborn lambs rapidly induces the disease in 3–6 2. OPA: A VIRUS-INDUCED weeks [57]. In natural conditions, the rapid CANCER development of tumoral lesions in young animals suggests a greater susceptibility of 2.1. Natural history the developing lung to JSRV [8]. In utero transmission to the fetus has been sug- Transmission of OPA among sheep has gested [8]. been suspected for almost two centuries. The first report dates back to 1825, with a letter written by a farmer who complained 2.2. JSRV: a member of the about the loss of many of his sheep. The Betaretrovirus genus disease was called “Jaagsiekte”, after the Afrikaans words for “chase” (Jaag) and JSRV is the virus responsible for the “sickness” (sieckte), to describe the respi- induction of OPA. JSRV belongs to the ratory distress observed in an animal out family Retroviridae, to the subfamily Or- of breath from being chased [91]. The first thoretrovirinae and the genus Betaretro- evidence of a viral cause came from the virus.TheBetaretrovirus genus also com- observation of retrovirus particles in the prises Mouse Mammary Tumour Virus lungs from sheep presenting clinical signs (MMTV) responsible for a mammary ade- of cancer [66], and was further clearly con- nocarcinoma in the mouse, Mason-Pfizer firmed by experimental induction of the Monkey Virus (MPMV) isolated from disease by intratracheal inoculation of viral a Rhesus monkey, and Squirrel Monkey particles with a activ- RetroVirus (SMRV). In 2003, Dolly the ity [47], cytoplasmic fractions of tumoral sheep, the first mammal cloned from an cells [82–84] or pulmonary secretions [76]. adult cell, died after being diagnosed with JSRV and lung cancer in sheep 213

Figure 1. Organisation (A) and transcription pattern (B) of JSRV provirus (from [30]). The gag, pro, and genes respectively encode the core proteins, the protease, the enzymatic activities and the envelope . “x” is an additional open reading frame encoding a putative protein of unknown function. LTR: . SA: splice acceptor; SD: splice donor (adapted from [80]). an incurable pulmonary adenocarcinoma the host-cell membrane (lipid bilayer and induced by JSRV. proteins). The virions carry two copies of The first attempts to characterise the the genome, composed of linear, positive, virus were made in the early 1980’s by single-stranded RNA. The 7.58 kb genome purification of the virus from lung lavage of the infectious virus comprises four main [91]. In 1991, a cDNA library was obtained genes, organised as 5’- gag- pro- pol- env - from purified 1.186 g/mL density fractions 3’ (Fig. 1), which encode the virus proteins [89], thus allowing the first sequencing of (reviewed in [64, 90]). Interestingly, while the entire JSRV genome of a South African the nucleotide sequences of gag, pro and isolate [90]. pol are homologous to their counterparts in MPMV, the env gene of JSRV is more related to that of MMTV and Human En- 2.3. Virus and genomic organisation dogenous Retrovirus -K (HERV-K). JSRV is organised as a simple retrovirus, with are RNA infect- an additional open reading frame, named ing vertebrate species and many non- ORF-x, that overlaps the 3’ end of the pol vertebrates. Virions (80–100 nm in diam- gene. ORF-x is unique among retroviruses eter) are spherical and surrounded by an and may encode a putative accessory pro- envelope, with spikes composed of virus- tein of 166 amino acids. Although the encoded glycoproteins. The envelope is existence of the protein is debated, ORF-x composed of viral proteins and elements of is strongly conserved in various isolates, 214 C. Leroux et al. and sequence analyses suggest a selective lipid bilayer and are composed of a hy- pressure for its conservation [6, 63, 72, 91] drophobic stretch of amino acids followed (and our unpublished data). Interestingly, by a short cytoplasmic tail. The JSRV en- two sub-genomic mRNA with splice ac- velope is a major determinant for the cel- ceptor sites within or in the vicinity of lular transformation as discussed in detail ORF-x have been identified, suggesting below. that this putative gene may actually be tran- JSRV is phylogenetically related to the scribed [63]. We have also identified these Enzootic Nasal Tumour Virus (ENTV), two mRNA in tumoral lungs (unpublished the agent responsible for nasal adenocarci- data). noma, a contagious tumour of the mucosal The JRSV genome contains non-coding nasal glands affecting sheep and goats. In regions at the ends of the genome, that are infected animals, epithelial cell prolifer- essential for the virus replication: R re- ation is responsible for continuous nasal peated at both ends; U5 unique to the 5’ discharge, respiratory distress, exophthal- end and U3 unique to the 3’ end. The inte- mos and important skull deformations (for grated genome is flanked by the long termi- review see [19]). Co-infection of ENTV nal repeat (LTR) composed of the 3 regions and JSRV has been reported [54]. U3-R-U5. The LTR serve as the sites of A family of endogenous retroviruses, transcriptional initiation. The U3 region enJSRV (endogenous JSRV) closely re- contains several elements important for vi- lated to JSRV, is present in domestic and ral transcription and tropism. The gag gene wild sheep and goats [34]. JSRV and en- (group-specific- antigen) encodes a single JSRV are highly related with polyprotein that is cleaved into at least 90–98% homology in deduced amino-acid three proteins: the matrix (MA), the major sequences [59]. Endogenous retroviruses capsid protein (CA) and the nucleocap- (ERV) are vertically transmitted as sta- sid (NC). The pro gene encodes a protein ble Mendelian genes in the germline of compatible with already-described dUT- most . They derive from the in- Pase in its 5’ part and a protease (PRO) tegration of exogenous viruses in the host in its 3’ end. Whereas the protease cleaves genomes, followed by genetic stabilisation the precursor polyproteins, the dUTPase through accumulation of mutations. Sev- (reviewed in [11]) prevents the incorpora- eral families of HERV such as HERV-F, tion of deoxyuridine triphosphate (dUTP) HERV-FRD, HERV-K, HERV-R, HERV-T by the reverse transcriptase [91]. The pol and HERV-W (defined by the tRNA com- gene is predicted to encode the enzymatic plementary to their putative primer binding activities: Reverse Transcriptase (RT) and site using the one letter code for the tRNA’s integrase (IN), respectively implicated in corresponding ) have been de- the replication of the viral RNA and in scribed in the human genome. Eight to the integration of the retrotranscribed DNA twelve copies of enJSRV have been lo- provirus into the host genome. The RT is a cated on metaphase chromosome spreads RNA-dependent DNA polymerase, essen- of sheep and goats using fluorescent in situ tial for the conversion of viral RNA into hybridisation [10] (Fig. 2). DNA. The env gene encodes the surface The biological significance of ERV is (SU) and transmembrane (TM) glycopro- still largely debated. Expression of ERV teins. The SU glycoprotein interacts with has been described in the placenta and gen- the cellular receptor of JSRV in sheep, ital tract of mammals including humans Hyal-2, a glycosylphosphatidylinositol an- with HERV-W. In the female genital tract, chored hyaluronidase-2 [70]. The TM gly- enJSRV expression is limited to the ep- coproteins presumably anchor SU into the ithelia and is particularly abundant in the JSRV and lung cancer in sheep 215

cellular tropism. Hence, ovine cell lines issued from various tissues may be in- fected in vitro by JSRV, however viral production stays weak [58]. Presence of vi- ral DNA can be demonstrated in naturally infected animals in lymphoid tissues, alve- olar macrophages and in peripheral blood mononuclear cells such as monocytes or B and T lymphocytes [29, 33, 37, 56, 75]. Viral envelope and LTR regions are es- sential determinants for the tropism and the expression of retroviruses. Located at the surface of the viral particle, sur- face (SU) glycoproteins specifically inter- Figure 2. Chromosomal localisation of the en- act with cellular receptors, allowing entry JSRV copies. Integration sites were determined of the virus into the cell. It is now well es- by in situ hybridisation with a full-length JSRV tablished that Hyal2 (hyaluronidase 2) is probe on ovine metaphase spreads. Chromo- the cellular receptor for JSRV (Fig. 3) in somes were identified by R-banding, and in- sheep [23, 70]. Hyal2 is a member of the tegration sites are indicated by arrows. Chr.: hyaluronglucosaminidase family, enzymes Chromosome. (A color version of this figure is that degrade hyaluronic acids of the ver- available at www.edpsciences.org/vetres.) tebrates’ extracellular matrix. Hyal2 only shows a weak hyaluronidase activity in comparison to other proteins of the same endometrium [22,25,26,62].Envelope pro- family. Hyal2 a glycosylphosphatidylinos- teins and env mRNA are present in the itol (GPI)-anchored cell-surface receptor, mononuclear trophoblasts and particularly is ubiquitously expressed in human and abundant in the binucleate cells and the mouse tissues [14]. Hyal2 also acts as syncytiotrophoblast plaques of the ovine a cellular receptor for ENTV [23], the placenta [24,25,62]. Only low levels of ex- related Betaretrovirus that induces nasal pression have been described in the lung tumours. Ubiquitous expression of Hyal2 epithelium [62]. is expected in sheep, in accordance with the capacity of JSRV to infect different 2.4. JSRV tropism cell types. JSRV may thus be able to en- ter different cells via its ubiquitous re- JSRV is unique among the retroviruses ceptor [56, 58], however its active repli- in that it induces transformation of dif- cation is restricted to bronchioloalveolar ferentiated lung epithelial cells, i.e. type epithelial cells (tropism restriction). In- II pneumocytes (also called alveolar type deed, the productive infection is strictly II cells) in the alveoli, and Clara cells in controlled at the cellular and molecular the bronchioli. Tumours exclusively occur level. Retroviral LTR contain the viral pro- in the sheep lung, as a result of selec- moter and enhancer elements that inter- tive replication of JSRV in these two cell act with cellular transcription factors; they types demonstrated by the detection of the are specifically activated in cells express- only in the tumoural cells ing transcription factors that bind to the and their neighbouring epithelial cells [55, enhancer regions. Restriction of JSRV ex- 68, 75]. However JSRV may infect dif- pression to lung epithelial cells is largely ferent cell types in vitro, with a larger due to its LTR transcriptional specificity 216 C. Leroux et al.

Figure 3. Pathways involved in JSRV-induced transformation and in the maintenance of the tu- mour (adapted from [80]). Two main pathways have been shown to be activated following in vitro expression of the JSRV envelope in different cell lines: the MAPKinase and PI3K/Akt pathways.

[60]. Promoters of specific genes from The endogenous virus enJSRV uses the type II pneumocytes or Clara cells, such same Hyal2-cellular receptor as JSRV. This as surfactant proteins SP-A, SP-B, SP-C, could lead to a phenomenon of interference SP-D and CCSP (Clara Cell Secretory Pro- at the time of cell infection by the exoge- tein or CC10) share different regulatory nous form of the virus [79]. Exogenous elements. JSRV LTR have several bind- JSRV and enJSRV are differentially regu- ing sites for transcription factors. Among lated, since LTR of enJSRV may respond those, HNF-3 (Hepatocyte Nuclear Factor to progesterone but not to the transcription 3) a factor involved in regulating the ex- factors that regulate expression of exoge- pression of surfactant protein genes, and nous virus LTR [59]. C/EBP (CCAAT/Enhancer Binding Pro- tein), are essential for the transcriptional activity of JSRV LTR. Directed mutagene- 3. THE JSRV INDUCED DISEASE sis experiments have established that alter- 3.1. Clinical presentation and β / ations of NF-I, HNF3 and C EBP binding histopathology sites reduce LTR activity of JSRV by 40 to 70% in MLE-15, a murine type II pneu- OPA shows different symptoms such mocyte cell line [48, 60]. C/EBP is also an as progressive dyspnea, abundant bron- important activator of transcriptional activ- chorrhea, cough, anorexia and cachexia. ity of the LTR in MtCC1-2, a murine Clara Death usually results from end-stage respi- cell line, while HNF-3 is not essential [48]. ratory failure. A typical sign (known as the JSRV and lung cancer in sheep 217

Figure 4. Subtypes of lesions observed in OPA: bronchioloalveolar (A), acinar (B) and papillary (C). (A color version of this figure is available at www.edpsciences.org/vetres.)

“wheelbarrow test”) is the flow of abun- these three histopathological subtypes may dant mucoid fluid (up to 500 mL per day in be present within the same tumoural tissue. advanced disease) from nostrils when the The OPA tumoural cells derive from rear legs of the animal are raised. Extra- epithelial cells of the distal lung, namely thoracic metastases are rare. Macroscopic alveolar type II cells (for ∼80% of the examination shows enlarged lungs infil- cells) and Clara cells (for ∼15% of the trated with tumoural areas, varying from cells) [68] (Fig. 5). Type II pneumocytes small nodules (1–30 mm) to lobar consol- produce different components of surfac- idation [18, 51]. The disease is multifocal, tant, a tensio-active agent that allows the disseminated in both lungs, pneumonic in maintenance of alveolar integrity. appearance, with the airways filled with Interestingly, tumoural cells over- fluid produced by the tumoural cells. express CD208/DC-LAMP (Dendritic Cell Lyzosomal Associated Membrane According to the latest WHO classifi- Protein) [74], a protein belonging to the cation for human lung cancers [7], OPA LAMP family of proteins (Lysosomal- should be referred to as a mixed adeno- Associated Membrane Protein), and carcinoma with associated bronchioloalve- initially described in activated dendritic olar, papillary and/or acinar subtypes. The cells. CD208/DC-LAMP is also con- subtypes are defined by their stitutively expressed in human, murine and differentiation patterns [51] (Fig. 4). and ovine type II pneumocytes, and is The bronchioloalveolar differentiation is over-expressed in human bronchioloalve- characterised by the expansion of cells fol- olar carcinoma and OPA [74]. In type II lowing the alveolar septa, also referred pneumocytes, CD208 is expressed in the to as the lepidic spread, without destruc- constitutive membranes of the lamellar tion of the alveolar architecture. The pap- bodies, specific vesicles specialised in illary subtype is defined by the pres- surfactant production. The role of CD208 ence of papilla-like structures protruding over-expression in tumoural development above the epithelial layer and replacing is still unknown. the underlying alveolar architecture. Fi- nally, the acinar subtype is composed of 3.2. Mechanisms of oncogenesis duct-like structures or acini and tubules composed of cells resembling bronchial Oncogenic retroviruses have been glands. In JSRV-induced adenocarcinoma, historically divided into two groups 218 C. Leroux et al.

Figure 5. Microscopic pattern of lesions from OPA. Typical aspect of a non-tumoural (A) and tumoural ovine lung (B) labelled with an anti SP-A (Surfactant Protein A) antibody. Arrows show tumoural cells, i.e. type II pneumocytes labelled with and anti SP-A antibody. (A color version of this figure is available at www.edpsciences.org/vetres.) depending on the speed of disease induc- not present any sequence homology with tion. Acute and non acute retroviruses known [6, 72], and its deletion cause tumours with a short or long does not prevent transformation in vitro. latency period respectively. Acute retro- It is now clearly established that JSRV viruses have been described in mice, induces tumours via the oncogenic prop- birds, and are oncogene-bearing erties of its envelope, which is both nec- retroviruses. Viral oncogenes are derived essary and sufficient to induce transfor- from normal cellular genes or proto- mation. The transforming property of the oncogenes, involved in regulation of cell JSRV envelope has been demonstrated in proliferation. Once captured by the virus, vitro in various cell lines including murine proto-oncogenes undergo mutations that NIH 3T3 fibroblasts [44], rat 208F fibrob- lead to uncontrolled cell proliferation. lasts [70], avian DF-1 fibroblasts [4, 93], (RSV) is an example bronchial human BEAS-2B epithelial cells of avian retrovirus bearing an oncogene, [16], canine kidney MDCK epithelial cells v-src, deriving from a mammalian cellular [43], and rat kidney RK3E cells [46]. kinase. The oncogenic property of the JSRV en- Typical lesions of OPA may be observed velopehasalsobeenshowninvivoinan in just a few weeks following experimental immuno-deficient mouse model [87] and inoculation of concentrated JSRV particles recently in sheep [9]. Expression of the [64, 83, 84]. This timeframe of disease in- JSRV envelope in mice using a replication- duction and the multifocal pattern of OPA incompetent adeno-associated virus vector, are compatible with the presence of an AAV6, results in lung tumours similar to oncogene in the JSRV genome [18]. JSRV those observed in sheep [87]. The bron- DNA transfection in murine NIH 3T3 fi- chioloalveolar localisation of the tumours broblasts induces foci of transformed cells, and the expression of the surfactant protein clearly indicating that the JSRV genome SP-C show that the transformed cells are contains an oncogenic element [44, 70]. derived from type II pneumocytes [87]. Us- However, to date no sequence homolo- ing a replication-defective virus carrying gous to any cellular oncogene has been the env gene under the control of the JSRV detected in the JSRV genome [72]. The LTR, it has been shown that the JSRV open reading frame (orf-x) of JSRV might envelope is sufficient to induce lung tu- be a potential oncogene, although it does mours in sheep. The envelope of the related JSRV and lung cancer in sheep 219

ENTV virus displays the same oncogenic RK3E cells [38]. Akt activation was ob- property both in vitro [2, 23] and in vivo served in different cell lines transformed [88]. by JSRV (Fig. 3), while it was absent from Deletion experiments show that the TM parental cells. OPA-derived type II pneu- (transmembrane) region of the envelope mocytes do not respond to EGF (Epidermal is the main determinant for cell trans- Growth Factor) stimulation, an activator formation [12, 36, 38]. In addition to the of the PI3K-Akt pathway [80], suggest- TM region, deletions of the SU (sur- ing dysregulation of the Akt pathway by face) glycoprotein (going from the signal JSRV infection. We recently showed that peptide to the junction between the SU Akt activation was observed in 37% of and TM subunits) also abolish transfor- OPA tumours sampled at a late stage of mation induced by the envelope, suggest- disease [80]. These results support the in- ing that multiple domains of SU may be volvement of the Akt signalling pathway in involved in cellular transformation. The the development and/or the maintenance of cytoplasmic tail of TM, composed of 43 OPA, and also suggest the involvement of amino acids, is essential for the trans- an Akt-independent pathway [80]. formation process in MDCK and NIH- The mechanisms leading to JSRV- 3T3 cells [43, 61]. This region contains induced cell transformation are in fact a peptidic YXXM motif (Fig. 3), cor- much more complex than previously con- responding to a potential consensus site sidered. Several experiments rule out a (phosphorylated on tyrosine Y) linked to direct role for the YXXM motif in Akt ac- the SH2 domain of the p85 subunit of tivation. In JSRV-transformed cells, phos- PI3K (Phosphatidyl-Inositol-3 Kinase), a phorylation of the Y590 residue of the kinase that activates Akt. The PI3K-Akt intra-cytoplasmic TM tail, or interaction signalling pathway is determinant in cel- between the p85 subunit of PI3K and the lular proliferation and survival (for review envelope (a prerequisite for Akt activa- [15]) (Fig. 3). Following a membrane stim- tion), have never been shown [42, 43]. ulus such as a growth factor binding to its Mutations of the YXXM motif do not abol- receptor, PI3K is recruited to the cell mem- ish Akt phosphorylation, so that the exact brane. Phosphorylated-PI3K phosphory- role of this motif remains to be deter- lates the second messenger PIP2 (phos- mined [42, 43, 93]. Moreover, implication phatidylinositol (4,5) biphosphate) into of the YXXM motif may differ between active PIP3 (phosphatidylinositol (3,4,5) cell types in in vitro experiments [4, 36, triphosphate). PIP3 can then recruit PDK1 41, 42, 61, 93]; while it is essential for (phosphatidylinositol-dependent kinase 1), transformation of NIH 3T3 fibroblasts, it which in turn phosphorylates Akt. The ki- is not required for transformation of avian nase Akt can phosphorylate diverse sub- DF-1 fibroblasts. Nevertheless, the pres- strates involved in signalling cascades con- ence of the YXXM motif seems to affect trolling cellular proliferation, survival and the efficiency of envelope-mediated trans- metabolism. Akt phosphorylation inhibits formation [43]. Since the YXXM motif proteins such as GSK-3 (Glycogen Syn- does not explain the oncogenic properties thase Kinase 3), FOXO (forkhead box tran- of JSRV, the mechanism of Akt activation scription factor), p24 or Kip1, and activates in transformed cells still need to be de- mTOR (mammalian target of rapamycin), termined. Interestingly, treatment of cells an important regulator of cell growth. with LY294002, a PI3K-specific inhibitor, Mutations of the YXXM motif of the drastically reduces Akt phosphorylation in JSRV-env gene abolish cell transformation NIH 3T3 cells [42-45, 93], suggesting that of NIH-3T3 [43, 61] and rat 208F and PI3K-dependent Akt activation may occur. 220 C. Leroux et al.

Figure 6. Activation of PI3K/Akt and MAPK pathways. Binding of growth factor, such as epi- dermal growth factor (EGF) to its receptor (such as EGF receptor) may induce activation of different pathways involved in the control of cell proliferation, apoptosis and differen- tiation (adapted from [80]).

In addition, PI3K inactivation does not MAPK are regulated and phosphorylated prevent cell transformation [46], and Akt by the MAPK kinase (MAPKK) such activation may be observed in NIH 3T3 as MEK1/2, themselves activated by the cells deficient for the p85 subunit of PI3K MAPKK kinase (MAPKKK) such as Raf [45], suggesting that PI3K may not be nec- (Fig. 6). Activation of this signalling cas- essary for Akt activation [28, 73]. cade leads to translocation of MAPK into JSRV-mediated cell transformation is the nucleus, where they activate differ- associated with the activation of the Ras- ent transcription factors. The Ras-MEK- MEK-MAPK pathway. Mitogen-activated MAPK pathway is activated in JSRV- Protein Kinase (MAPK) are a group transformed NIH 3T3 and RK3E cells of kinases important in signal transduc- [38, 46] (Fig. 3). As we have reviewed tion, that may be activated by the small above for Akt, the significance of this Ras protein (Fig. 6). The MAPK path- signalling pathway in cell transformation way includes 4 protein families: ERK1/2 seems to vary according to the cell types (extracellular signal-regulated kinase 1 used in the in vitro studies [46]. Inhibitors and 2) also named p44/42; SAPK/JNK of MEK1 (MAPKK), can totally abolish (stress-activated protein kinase/c-Jun NH2- transformation in fibroblasts and epithe- terminal kinase); MAPK p38 and BMK lial cells. On the contrary, although an (big mitogen-activated protein kinase 1). inhibitor of MAPKKK totally abolishes the JSRV and lung cancer in sheep 221

Figure 7. Tumoural type II pneumocytes in OPA-derived primary cultures. Cytoplasm is rich in lamellar bodies (LB) (A) as shown in an enlarged view (B). In vitro-derived cells express cytokeratin (C), the surfactant protein C (SP-C) (D) and co-express both cytokeratin and SP-C (E). (A color version of this figure is available at www.edpsciences.org/vetres.) transformation of fibroblast NIH 3T3, its lungs [5, 80]; those tumour-derived cells effect on epithelial RK3E cells is only par- are clearly type II pneumocytes as revealed tial. A recent study on JSRV and ENTV by the presence of lamellar bodies in the tumoural tissues gives more clues to un- cytoplasm, and the expression of surfac- derstand the JSRV complex signalling cas- tant protein C (SP-C) (Fig. 7). Analysis cades. In naturally and experimentally in- of transformation mechanisms in the ovine duced OPA, MAPK Erk1/2 seems to be the type II pneumocytes is in progress. predominant activated pathway [20]. The activation of telomerase was re- Since the first description of the onco- cently evidenced in OPA-derived type II genic properties of the JSRV envelope, pneumocytes and in tumoural lung tissues, many studies performed in various cell suggesting that replicative senescence may types (fibroblasts, epithelial cells) and be negatively regulated in this tumour [80] species (human, sheep, mouse, rat) have (Fig. 3). Replicative senescence, leading to led to a better understanding of the mech- the natural death of the cells after several anisms leading to cell transformation. divisions, is essential for the maintenance Although a number of points remain unan- of homeostasis and strict regulation of the swered, the transformation steps are be- cell life span. Replicative senescence is lieved to be dependent on the nature and mainly regulated by the telomerase, a ri- origin of the cells. Hence, understanding bonucleoprotein enzyme complex able to the key events in the transformation of maintain telomere length during cell divi- ovine type II pneumocytes (i.e. cells that sion, by de novo synthesis of telomeres and represent the cells at the origin of the tu- elongation of existing telomeres. Telom- mours) is now the challenge of ongoing erase activation has been implicated as work. Our group has developed primary a crucial factor in oncogenesis through cell cultures isolated from tumoural ovine inhibition of replicative senescence and 222 C. Leroux et al. down-regulation of cell death. Telomerase receptor tyrosine kinase Stk/Ron [40, 53] activity is itself regulated by a variety of that induces the proliferation of the tar- factors, including the Akt kinase pathway. get cell. Despite the direct effect of the We reported telomerase activation and Akt envelope on cell signalling, other molecu- activation in OPA tumours [80], suggest- lar events are necessary for cell transfor- ing that Akt activation may contribute mation, such as integration of the virus to telomerase activation and inhibition of genome in the vicinity of protooncogenes senescence and cell death in JSRV infected [71]. cells, thereby contributing to the accumu- Tumorigenesis is a multistep process lation of tumoural cells in the ovine lung. and the JSRV envelope might not be suf- In most of the cell lines studied, JSRV ficient to induce transformation of cells envelope-induced transformation is inde- in vivo. Insertional mutagenesis cannot be pendent of its interaction with the cellular ruled out. Except in neonates, OPA is char- receptor Hyal2. Hyal2 is weakly active as a acterised by a slow progression from in- JSRV receptor in mice and the deletion of fection to disease, suggesting a non-acute the receptor binding domain (RBD), pre- mechanism of transformation [78]. A multi dicted by sequence homology with other step gene walking technique was recently retroviruses, from the JSRV envelope does used to clone and sequence JSRV inte- not abolish the transformation [41, 49, 50, gration sites from sheep with OPA [13] 70]. and identified multiple integration sites in Although incompletely understood, the most tumours, suggesting a random distri- original mechanism of JSRV-cell transfor- bution. Interestingly, a common integration mation may not be unique among retro- site has been described in sheep chromo- viruses. Envelope-mediated activation of some 16 [13] and in or near the receptor cellular proliferation has also been shown protein tyrosine phosphatase γ (RPT Pase for MMTV [39], Spleen Focus Forming γ) gene [67]. In this context, insertional Virus (SFFV) [1, 86] and Avian Heman- mutagenesis may participate to the devel- gioma Virus (AHV) [3]. Expression of opment of the tumour in sheep. the MMTV envelope in the human and murine mammary epithelial cell line in- duces morphological changes compatible 4. JSRV AND HUMAN LUNG with cell transformation [39]; cell trans- CANCER formation is dependent on an Immunore- ceptor Tyrosine-based Activation Motif Fifteen to 20% of all human cancers (ITAM motif), a potential anchor site for are thought to be associated with infec- signalling proteins carrying a SH2 motif, tious agents (reviewed in [31]). OPA has and encoded by the MMTV env gene [39]. been considered as a model for human ade- ITAM (Yxx(L/I)x6−8Yxx(L/I)) motifs are nocarcinomas, and especially pneumonic- associated with cell survival, activation type bronchioloalveolar carcinoma (BAC), and differentiation; tyrosine (Y) residues since the two tumours share common clini- are necessary and sufficient for signalling cal, radiological and histopathological fea- function. The envelope-mediated transfor- tures [51]. Similarly to OPA, BAC is a mation has been well established in the slow-growing tumour with rare metastatic case of SFFV, a murine retrovirus caus- spread. It is clinically associated with ing erythroleukemia. The SFFV genome highly productive cough and progressive encodes a truncated envelope protein that restrictive respiratory failure [51, 85]. The interacts with and activates the erythropoi- epidemiology of BAC differs from that of etin receptor, and a truncated form of the other non-small cell lung cancers, with a JSRV and lung cancer in sheep 223

Table I. JSRV and human lung cancer.

For Against Analogy to the ovine pulmonary adenocarci- noma Specific epidemiology of the bronchioloalveo- No epidemiologic evidence of JSRV transmission lar cancer from sheep to humans Presence of JSRV-receptor Hyal2 in human Lack of JSRV DNA or RNA in human adenocarci- cells noma Possible neoplastic transformation of human cells with JSRV envelope protein Evidence for a protein related immunologically to JSRV in some human lung tumours Detection of JSRV-related DNA sequences in Africans less important epidemiologic link to to- servations suggest the existence of either bacco smoking, an increased frequency in extra-pulmonary tumoural stem cells that women and younger patients, and a bet- would remain dormant for several years ter outcome than other non-small cell lung [32], or of infectious agents with an extra- cancers (with a 5-year survival of about pulmonary preclinical reservoir. 60%). Given the similarities of OPA and Hyal2, the cellular receptor of JSRV, has human BAC, a viral cause to human BAC been shown to be present at the surface has long been hypothesised; several recent of a wide range of human cells, includ- clinical and molecular observations have ing alveolar cells, and moreover to bind been reported studying the link between the JSRV envelope protein [70]. Further- JSRV and human lung carcinoma (Tab. I). more, the gene coding for Hyal2 is located Clinically, an interesting argument in in the chromosomal region 3p21 which is favour of an underlying infectious condi- frequently deleted in human lung cancer, tion in human lung cancer came from the making this gene a potential tumour sup- pattern of recurrence of BAC after lung pressor in lung [69, 70]. transplantation. Although usually con- traindicated in cancer, lung transplantation Interestingly, de las Heras et al. [17] has been reported as a feasible treatment showed the presence of an antigen cross- of unresectable BAC because of the lack reacting with JSRV-Gag antiserum in about of metastatis in this tumour type. Since 30% of human BAC, and 26% of lung ade- nocarcinomas. However, these results have our first report of lung transplantation in a patient with BAC [27], about thirty cases not been confirmed by further molecular have been described, showing recurrence studies and 2 PCR-based analyses failed rates of about 50%, within a median time to detect any exogenous or endogenous JSRV-related genome in human BAC and of 45 months [21]. Microsatellite analy- sis of the lung specimens of the donor adenocarcinoma [35, 92]. and the recipient have showed that recur- More recently, Morozov et al. [52] re- rent tumours originated from the transplant ported the presence of JSRV-related se- recipient [30, 65]. Since BAC is consid- quences in healthy and HIV positive ered as a localised disease (and excluding Africans, but not in the few lung cancer pa- possible tumoural contamination during tients tested. Their significance remains to the transplantation procedure), these ob- be evaluated. 224 C. Leroux et al.

Up to date, no conclusive molecular ev- [4] Allen T.E., Sherrill K.J., Crispell S.M., idence of a link between JSRV and human Perrott M.R., Carlson J.O., DeMartini J.C., The jaagsiekte sheep retrovirus envelope BAC has been provided (Tab. I). As an gene induces transformation of the avian fi- alternative approach, a case control epi- broblast cell line DF-1 but does not require demiologic study is currently underway in a conserved SH2 binding domain, J. Gen. France, conducted by our group. The ob- Virol. (2002) 83:2733–2742. jective is to determine if ovine or caprine [5] Archer F., Jacquier E., Lyon M., Chastang J., exposure is a risk factor for BAC in hu- Cottin V., Mornex J.-F., Leroux C., Alveolar Type II cells isolated from pulmonary adeno- mans. carcinoma: a model for JSRV expression in vitro, Am. J. Respir. Cell. Mol. Biol. (2006) (in press). 5. CONCLUSION [6] Bai J., Bishop J.V., Carlson J.O., DeMartini J.C., Sequence comparison of JSRV with Over the last few years, research on endogenous proviruses: envelope genotypes JSRV and the resulting OPA has focussed and a novel ORF with similarity to a G- on the molecular mechanisms of cell trans- protein-coupled receptor, (1999) formation. Interesting results came from 258:333–343. the discovery that the virus envelope acts [7] Beasley M.B., Brambilla E., Travis W.D., The 2004 World Health Organization classi- as a potent oncogene. Several cellular fication of lung tumors, Semin. Roentgenol. pathways seem to occur in the cell trans- (2005) 40:90–97. formation. [8] Caporale M., Centorame P., Giovannini A., To conclude, research into the biology Sacchini F., Di Ventura M., De las Heras of JSRV and mechanisms leading to the de- M., Palmarini M., Infection of lung epithelial velopment of OPA is of great interest both cells and induction of pulmonary adenocar- cinoma is not the most common outcome for the naturally induced cancer in sheep of naturally occurring JSRV infection during and for BAC, the related human cancer. the commercial lifespan of sheep, Virology Even though a viral agent remains uncer- (2005) 338:144–153. tain in BAC patients, understanding the [9] Caporale M., Cousens C., Centorame P., steps leading to the transformation of lung Pinoni C., De las Heras M., Palmarini M., epithelia may be of interest in the context Expression of the jaagsiekte sheep retro- virus envelope glycoprotein is sufficient to of therapeutic approaches in human lung induce lung tumors in sheep, J. Virol. (2006) cancers in general and BAC in particular. 80:8030–8037. [10] Carlson J., Lyon M., Bishop J., Vaiman A., Cribiu E., Mornex J.F., Brown S., Knudson REFERENCES D., DeMartini J., Leroux C., Chromosomal distribution of endogenous Jaagsiekte sheep [1] Aizawa S., Suda Y., Furuta Y., Yagi T., retrovirus proviral sequences in the sheep Takeda N., Watanabe N., Nagayoshi M., genome, J. Virol. (2003) 77:9662–9668. Ikawa Y., Env-derived gp55 gene of Friend spleen focus-forming virus specifically in- [11] Chen R., Wang H., Mansky L.M., Roles duces neoplastic proliferation of erythroid of uracil-DNA glycosylase and dUTPase progenitor cells, EMBO J. (1990) 9:2107– in virus replication, J. Gen. Virol. (2002) 2116. 83:2339–2345. [2] Alberti A., Murgia C., Liu S.L., Mura [12] Chow Y.H., Alberti A., Mura M., Pretto C., M., Cousens C., Sharp M., Miller A.D., Murcia P., Albritton L.M., Palmarini M., Palmarini M., Envelope-induced cell trans- Transformation of rodent fibroblasts by the formation by ovine , J. Virol. jaagsiekte sheep retrovirus envelope is re- (2002) 76:5387–5394. ceptor independent and does not require the surface domain, J. Virol. (2003) 77:6341– [3] Alian A., Sela-Donenfeld D., Panet A., Eldor 6350. A., Avian hemangioma retrovirus induces cell proliferation via the envelope (env) gene, [13] Cousens C., Bishop J.V., Philbey A.W., Gill Virology (2000) 276:161–168. C.A., Palmarini M., Carlson J.O., DeMartini JSRV and lung cancer in sheep 225

J.C., Sharp J.M., Analysis of integration sites and transformation by enzootic nasal tumor of Jaagsiekte sheep retrovirus in ovine pul- virus, J. Virol. (2002) 76:2141–2149. monary adenocarcinoma, J. Virol. (2004) [24] Dunlap K.A., Palmarini M., Adelson D.L., 78:8506–8512. Spencer T.E., Sheep endogenous betaretro- [14] Csoka A.B., Scherer S.W., Stern R., viruses (enJSRVs) and the hyaluronidase 2 Expression analysis of six paralogous (HYAL2) receptor in the ovine uterus and human hyaluronidase genes clustered on conceptus, Biol. Reprod. (2005) 73:271– chromosomes 3p21 and 7q31, Genomics 279. (1999) 60:356–361. [25] Dunlap K.A., Palmarini M., Spencer T.E., [15] Cully M., You H., Levine A.J., Mak T.W., Ovine endogenous betaretroviruses (enJS- Beyond PTEN mutations: the PI3K pathway RVs) and placental morphogenesis, Placenta as an integrator of multiple inputs during tu- (2006) 27 Suppl. A:S135–S140. morigenesis, Nat. Rev. Cancer (2006) 6:184– 192. [26] Dunlap K.A., Palmarini M., Varela M., Burghardt R.C., Hayashi K., Farmer J.L., [16] Danilkovitch-Miagkova A., Duh F.M., Spencer T.E., Endogenous retroviruses reg- Kuzmin I., Angeloni D., Liu S.L., Miller ulate periimplantation placental growth and A.D., Lerman M.I., Hyaluronidase 2 neg- differentiation, Proc. Natl. Acad. Sci. USA atively regulates RON receptor tyrosine (2006) 103:14390–14395. kinase and mediates transformation of epithelial cells by jaagsiekte sheep retro- [27] Etienne B., Bertocchi M., Gamondes J.P., virus, Proc. Natl. Acad. Sci. USA (2003) Wiesendanger T., Brune J., Mornex J.F., 100:4580–4585. Successful double-lung transplantation for bronchioalveolar carcinoma, Chest (1997) [17] De las Heras M., Barsky S.H., Hasleton 112:1423–1424. P., Wagner M., Larson E., Egan J., Ortin A., Gimenez-Mas J.A., Palmarini M., Sharp [28] Filippa N., Sable C.L., Filloux C., J.M., Evidence for a protein related immuno- Hemmings B., Van Obberghen E., logically to the jaagsiekte sheep retrovirus in Mechanism of protein kinase B activation by some human lung tumours, Eur. Respir. J. cyclic AMP-dependent protein kinase, Mol. (2000) 16:330–332. Cell. Biol. (1999) 19:4989–5000. [18] De las Heras M., Gonzalez L., Sharp J.M., [29] Garcia-Goti M., Gonzalez L., Cousens C., Pathology of ovine pulmonary adenocar- Cortabarria N., Extramiana A.B., Minguijon cinoma, Curr. Top. Microbiol. Immunol. E., Ortin A., De las Heras M., Sharp J.M., (2003) 275:25–54. Sheep pulmonary adenomatosis: characteri- [19] De las Heras M., Ortin A., Cousens C., zation of two pathological forms associated Minguijon E., Sharp J.M., Enzootic nasal with jaagsiekte retrovirus, J. Comp. Pathol. adenocarcinoma of sheep and goats, Curr. (2000) 122:55–65. Top. Microbiol. Immunol. (2003) 275:201– [30] Garver R.I. Jr., Zorn G.L., Wu X., McGiffin 223. D.C., Young K.R. Jr., Pinkard N.B., [20] De las Heras M.O.A., Benito A., Summers Recurrence of bronchioloalveolar carcinoma C., Ferrer L.M., Sharp J.M., In-situ demon- in transplanted lungs, N. Engl. J. Med. stration of mitogen-activated protein kinase (1999) 340:1071–1074. Erk 1/2 signalling pathway in contagious [31] Gatza M.L., Chandhasin C., Ducu R.I., respiratory tumours of sheep and goats, J. Marriott S.J., Impact of transforming viruses Comp. Pathol. (2006) 135:1–10. on cellular mutagenesis, genome stability, [21] De Perrot M., Chernenko S., Waddell T.K., and cellular transformation, Environ. Mol. Shargall Y., Pierre A.F., Hutcheon M., . (2005) 45:304–325. Keshavjee S., Role of lung transplantation [32] Gomez-Roman J.J., Del Valle C.E., in the treatment of bronchogenic carcinomas Zarrabeitia M.T., Martinez J.C., Goni for patients with end-stage pulmonary dis- F.Z., Lera R.M., Cuevas J., Val-Bernal J.F., ease, J. Clin. Oncol. (2004) 22:4351–4356. Recurrence of bronchioloalveolar carcinoma [22] DeMartini J.C., Carlson J.O., Leroux C., in donor lung after lung transplantation: mi- Spencer T., Palmarini M., Endogenous retro- crosatellite analysis demonstrates a recipient viruses related to jaagsiekte sheep retrovirus, origin, Pathol. Int. (2005) 55:580–584. Curr. Top. Microbiol. Immunol. (2003) [33] Gonzalez L., Garcia-Goti M., Cousens C., 275:117–137. Dewar P., Cortabarria N., Extramiana A.B., [23] Dirks C., Duh F.M., Rai S.K., Lerman Ortin A., De Las Heras M., Sharp J.M., M.I., Miller A.D., Mechanism of cell entry Jaagsiekte sheep retrovirus can be detected 226 C. Leroux et al.

in the peripheral blood during the pre- by jaagsiekte sheep retrovirus DNA, Proc. clinical period of sheep pulmonary adeno- Natl. Acad. Sci. USA (2001) 98:4449–4454. matosis, J. Gen. Virol. (2001) 82:1355–1358. [45] Maeda N., Inoshima Y., Fruman D.A., [34] Hecht S.J., Stedman K.E., Carlson J.O., Brachmann S.M., Fan H., Transformation DeMartini J.C., Distribution of endogenous of mouse fibroblasts by Jaagsiekte sheep type B and type D sheep retrovirus sequences retrovirus envelope does not require phos- in ungulates and other mammals, Proc. Natl. phatidylinositol 3-kinase, J. Virol. (2003) Acad. Sci. USA (1996) 93:3297–3302. 77:9951–9959. [35] Hiatt K.M., Highsmith W.E., Lack of DNA [46] Maeda N., Fu W., Ortin A., de las Heras evidence for jaagsiekte sheep retrovirus in M., Fan H., Roles of the Ras-MEK-mitogen- human bronchioloalveolar carcinoma, Hum. activated protein kinase and phosphatidyli- Pathol. (2002) 33:680. nositol 3-kinase-Akt-mTOR pathways in [36] Hofacre A., Fan H., Multiple domains of the Jaagsiekte sheep retrovirus-induced transfor- Jaagsiekte sheep retrovirus envelope protein mation of rodent fibroblast and epithelial cell are required for transformation of rodent fi- lines, J. Virol. (2005) 79:4440–4450. broblasts, J. Virol. (2004) 78:10479–10489. [47] Martin W.B., Scott F.M., Sharp J.M., [37] Holland M.J., Palmarini M., Garcia-Goti Angus K.W., Norval M., Experimental pro- M., Gonzalez L., McKendrick I., de las duction of sheep pulmonary adenomatosis Heras M., Sharp J.M., Jaagsiekte retrovirus (Jaagsiekte), Nature (1976) 264:183–185. is widely distributed both in T and B lym- [48] McGee-Estrada K., Fan H., In vivo and phocytes and in mononuclear phagocytes of in vitro analysis of factor binding sites in sheep with naturally and experimentally ac- Jaagsiekte sheep retrovirus long terminal re- quired pulmonary adenomatosis, J. Virol. peat enhancer sequences: roles of HNF-3, (1999) 73:4004–4008. NF-I, and C/EBP for activity in lung epithe- [38] Hull S., Fan H., Mutational analysis of lial cells, J. Virol. (2006) 80:332–341. the cytoplasmic tail of jaagsiekte sheep [49] Miller A.D., Identification of Hyal2 as retrovirus envelope protein, J. Virol. (2006) the cell-surface receptor for jaagsiekte 80:8069–8080. sheep retrovirus and ovine nasal adenocarci- [39] Katz E., Lareef M.H., Rassa J.C., Grande noma virus, Curr. Top. Microbiol. Immunol. S.M., King L.B., Russo J., Ross S.R., (2003) 275:179–199. Monroe J.G., MMTV Env encodes an ITAM [50] Miller A.D., Van Hoeven N.S., Liu S.L., responsible for transformation of mammary Transformation and scattering activities of epithelial cells in three-dimensional culture, the receptor tyrosine kinase RON/Stkinro- J. Exp. Med. (2005) 201:431–439. dent fibroblasts and lack of regulation by the [40] Li J.P., D’Andrea A.D., Lodish H.F., jaagsiekte sheep retrovirus receptor, Hyal2, Baltimore D., Activation of cell growth by BMC Cancer (2004) 4:64. binding of Friend spleen focus-forming virus [51] Mornex J.F., Thivolet F., De las Heras M., gp55 glycoprotein to the erythropoietin re- Leroux C., Pathology of human bronchi- ceptor, Nature (1990) 343:762–764. oloalveolar carcinoma and its relationship [41] Liu S.L., Duh F.M., Lerman M.I., Miller to the ovine disease, Curr. Top. Microbiol. A.D., Role of virus receptor Hyal2 in onco- Immunol. (2003) 275:225–248. genic transformation of rodent fibroblasts by [52] Morozov V.A., Lagaye S., Lower J., sheep betaretrovirus env proteins, J. Virol. Lower R., Detection and characterization (2003) 77:2850–2858. of betaretroviral sequences, related to [42] Liu S.L., Lerman M.I., Miller A.D., Putative sheep Jaagsiekte virus, in Africans from phosphatidylinositol 3-kinase (PI3K) bind- Nigeria and Cameroon, Virology (2004) ing motifs in ovine betaretrovirus Env pro- 327:162–168. teins are not essential for rodent fibroblast / [53] Nishigaki K., Thompson D., Hanson C., transformation and PI3K Akt activation, J. Yugawa T., Ruscetti S., The envelope glyco- Virol. (2003) 77:7924–7935. protein of friend spleen focus-forming virus [43] Liu S.L., Miller A.D., Transformation of covalently interacts with and constitutively madin-darby canine kidney epithelial cells activates a truncated form of the receptor ty- by sheep retrovirus envelope proteins, J. rosine kinase Stk, J. Virol. (2001) 75:7893– Virol. (2005) 79:927–933. 7903. [44] Maeda N., Palmarini M., Murgia C., Fan H., [54] Ortin A., Perez de Villarreal M., Minguijon Direct transformation of rodent fibroblasts E., Cousens C., Sharp J.M., De las Heras JSRV and lung cancer in sheep 227

M., Coexistence of enzootic nasal adenocar- [64] Palmarini M., Fan H., Molecular biology cinoma and jaagsiekte retrovirus infection in of jaagsiekte sheep retrovirus, Curr. Top. sheep, J. Comp. Pathol. (2004) 131:253–258. Microbiol. Immunol. (2003) 275:81–115. [55] Palmarini M., Dewar P., De las Heras [65] Paloyan E.B., Swinnen L.J., Montoya A., M., Inglis N.F., Dalziel R.G., Sharp J.M., Lonchyna V., Sullivan H.J., Garrity E., Epithelial tumour cells in the lungs of sheep Lung transplantation for advanced bronchi- with pulmonary adenomatosis are major oloalveolar carcinoma confined to the lungs, sites of replication for Jaagsiekte retrovirus, Transplantation (2000) 69: 2446–2448. J. Gen. Virol. (1995) 76:2731–2737. [66] Perk K., Michalides R., Spiegelman S., [56] Palmarini M., Holland M.J., Cousens C., Schlom J., Biochemical and morphologic Dalziel R.G., Sharp J.M., Jaagsiekte retro- evidence for the presence of an RNA tu- virus establishes a disseminated infection mor virus in pulmonary carcinoma of sheep of the lymphoid tissues of sheep affected (Jaagsiekte), J. Natl. Cancer Inst. (1974) by pulmonary adenomatosis, J. Gen. Virol. 53:131–135. (1996) 77:2991–2998. [67] Philbey A.W., Cousens C., Bishop J.V., Gill C.A., Demartini J.C., Sharp J.M., [57] Palmarini M., Sharp J.M., de las Heras Multiclonal pattern of Jaagsiekte sheep M., Fan H., Jaagsiekte sheep retrovirus is ffi retrovirus integration sites in ovine pul- necessary and su cient to induce a conta- monary adenocarcinoma, Virus Res. (2006) gious lung cancer in sheep, J. Virol. (1999) 117:254–263. 73:6964–6972. [68] Platt J.A., Kraipowich N., Villafane F., [58] Palmarini M., Sharp J.M., Lee C., Fan H., DeMartini J.C., Alveolar type II cells ex- In vitro infection of ovine cell lines by pressing jaagsiekte sheep retrovirus capsid Jaagsiekte sheep retrovirus, J. Virol. (1999) protein and surfactant proteins are the pre- 73: 10070–10078. dominant neoplastic cell type in ovine pul- [59] Palmarini M., Hallwirth C., York D., Murgia monary adenocarcinoma, Vet. Pathol. (2002) C., de Oliveira T., Spencer T., Fan H., 39:341–352. Molecular cloning and functional analysis [69] Rai S.K., DeMartini J.C., Miller A.D., of three type D endogenous retroviruses of Retrovirus vectors bearing jaagsiekte sheep sheep reveal a different cell tropism from that retrovirus Env transduce human cells by us- of the highly related exogenous jaagsiekte ing a new receptor localized to chromosome sheep retrovirus, J. Virol. (2000) 74:8065– 3p21.3, J. Virol. (2000) 74:4698–4704. 8076. [70] Rai S.K., Duh F.M., Vigdorovich V., [60] Palmarini M., Datta S., Omid R., Murgia Danilkovitch-Miagkova A., Lerman M.I., C., Fan H., The long terminal repeat of Miller A.D., Candidate tumor suppres- jaagsiekte sheep retrovirus is preferentially sor HYAL2 is a glycosylphosphatidylinosi- active in differentiated epithelial cells of the tol (GPI)-anchored cell-surface receptor for lungs, J. Virol. (2000) 74:5776–5787. jaagsiekte sheep retrovirus, the envelope pro- tein of which mediates oncogenic transfor- [61] Palmarini M., Maeda N., Murgia C., De- mation, Proc. Natl. Acad. Sci. USA (2001) Fraja C., Hofacre A., Fan H., A phos- 98:4443–4448. phatidylinositol 3-kinase docking site in the cytoplasmic tail of the jaagsiekte sheep retro- [71] Reboul J., Vaglio P., Rual J.F., Lamesch virus transmembrane protein is essential P., Martinez M., Armstrong C.M., Li S., for envelope-induced transformation of NIH Jacotot L., Bertin N., Janky R., Moore 3T3 cells, J. Virol. (2001) 75:11002–11009. T., Hudson J.R. Jr., Hartley J.L., Brasch M.A., Vandenhaute J., Boulton S., Endress [62] Palmarini M., Gray C.A., Carpenter K., Fan G.A., Jenna S., Chevet E., Papasotiropoulos H., Bazer F.W., Spencer T.E., Expression V., Tolias P.P., Ptacek J., Snyder M., of endogenous betaretroviruses in the ovine Huang R., Chance M.R., Lee H., Doucette- uterus: effects of neonatal age, estrous cycle, Stamm L., Hill D.E., Vidal M.C., Elegans pregnancy, and progesterone, J. Virol. (2001) ORFeome version 1.1: experimental veri- 75:11319–11327. fication of the genome annotation and re- [63] Palmarini M., Murgia C., Fan H., Spliced source for proteome-scale protein expres- and prematurely polyadenylated Jaagsiekte sion, Nat. Genet. (2003) 34:35–41. Sheep Retrovirus-specific from in- [72] Rosati S., Pittau M., Alberti A., Pozzi S., fected or transfected cells, Virology (2002) York D.F., Sharp J.M., Palmarini M., An 294:180–188. accessory open reading frame (orf-x) of 228 C. Leroux et al.

jaagsiekte sheep retrovirus is conserved be- transmission to lambs by means of cultured tween different virus isolates, Virus Res. cells and cell homogenates, Onderstepoort J. (2000) 66:109–116. Vet. Res. (1980) 47:13–18. [73] Sable C.L., Filippa N., Hemmings B., Van [84] Verwoerd D.W., Williamson A.L., De Obberghen E., cAMP stimulates protein Villiers E.M., Aetiology of jaagsiekte: kinase B in a Wortmannin-insensitive man- transmission by means of subcellular frac- ner, FEBS Lett. (1997) 409:253–257. tions and evidence for the involvement of a [74] Salaun B., de Saint-Vis B., Pacheco N., retrovirus, Onderstepoort J. Vet. Res. (1980) Pacheco Y., Riesler A., Isaac S., Leroux 47:275–280. C., Clair-Moninot V., Pin J.J., Griffith [85] Wislez M., Gounant V., Cadranel J., J., Treilleux I., Goddard S., Davoust J., / Bronchioloalveolar carcinoma, Rev. Mal. Kleijmeer M., Lebecque S., CD208 dendritic Respir. (2005) 22:8S70–8S75 (in French). cell-lysosomal associated membrane protein is a marker of normal and transformed [86] Wolff L., Ruscetti S., The spleen focus- type II pneumocytes, Am. J. Pathol. (2004) forming virus (SFFV) envelope gene, when 164:861–871. introduced into mice in the absence of other [75] Salvatori D., Gonzalez L., Dewar P., Cousens SFFV genes, induces acute erythroleukemia, C., de las Heras M., Dalziel R.G., Sharp J. Virol. (1988) 62:2158–2163. J.M., Successful induction of ovine pul- ff [87] Wootton S.K., Halbert C.L., Miller A.D., monary adenocarcinoma in lambs of di er- Sheep retrovirus structural protein induces ent ages and detection of viraemia during lung tumours, Nature (2005) 434:904–907. the preclinical period, J. Gen. Virol. (2004) 85:3319–3324. [88] Wootton S.K., Halbert C.L., Miller A.D., [76] Sharp J.M., Angus K.W., Gray E.W., Scott Envelope proteins of jaagsiekte sheep retro- F.M., Rapid transmission of sheep pul- virus and enzootic nasal tumor virus induce monary adenomatosis (jaagsiekte) in young similar bronchioalveolar tumors in lungs of lambs. Brief report, Arch. Virol. (1983) mice, J. Virol. (2006) 80:9322–9325. 78:89–95. [89] York D.F., Vigne R., Verwoerd D.W., [77] Sharp J.M., Angus K.W., Jassim F.A., Scott Querat G., Isolation, identification, and par- F.M., Experimental transmission of sheep tial cDNA cloning of genomic RNA of pulmonary adenomatosis to a goat, Vet. Rec. jaagsiekte retrovirus, the etiological agent (1986) 119:245. of sheep pulmonary adenomatosis, J. Virol. (1991) 65:5061–5067. [78] Sharp J.M., DeMartini J.C., Natural history of JSRV in sheep, Curr. Top. Microbiol. [90] York D.F., Vigne R., Verwoerd D.W., Querat Immunol. (2003) 275:55–79. G., Nucleotide sequence of the jaagsiekte [79] Spencer T.E., Mura M., Gray C.A., Griebel retrovirus, an exogenous and endogenous P.J., Palmarini M., Receptor usage and fetal type D and B retrovirus of sheep and goats, expression of ovine endogenous betaretro- J. Virol. (1992) 66:4930–4939. viruses: implications for coevolution of en- [91] York D.F., Querat G.A., A history of ovine dogenous and exogenous retroviruses, J. pulmonary adenocarcinoma (jaagsiekte) and Virol. (2003) 77:749–753. experiments leading to the deduction of [80] Suau F., Cottin V., Archer F., Croze S., the JSRV nucleotide sequence, Curr. Top. Chastang J., Cordier G., Thivolet-Bejui F., Microbiol. Immunol. (2003) 275:1–23. Mornex J.F., Leroux C., Telomerase activa- tion in a model of lung adenocarcinoma, Eur. [92] Yousem S.A., Finkelstein S.D., Swalsky Respir. J. (2006) 27:1175–1182. P.A., Bakker A., Ohori N.P., Absence of jaagsiekte sheep retrovirus DNA and RNA in [81] Tustin R.C., Williamson A.L., York D.F., bronchioloalveolar and conventional human Verwoerd D.W., Experimental transmission pulmonary adenocarcinoma by PCR and RT- of jaagsiekte (ovine pulmonary adenomato- PCR analysis, Hum. Pathol. (2001) 32:1039– sis) to goats, Onderstepoort J. Vet. Res. 1042. (1988) 55:27–32. [93] Zavala G., Pretto C., Chow Y.H., Jones L., [82] Verwoerd D.W., de Villiers E.M., On the ae- Alberti A., Grego E., De las Heras M., tiology of Jaagsiekte, J. S. Afr. Vet. Assoc. Palmarini M., Relevance of Akt phospho- (1980) 51:71–74. rylation in cell transformation induced by [83] Verwoerd D.W., De Villiers E.M., Tustin Jaagsiekte sheep retrovirus, Virology (2003) R.C., Aetiology of jaagsiekte: experimental 312:95–105.