Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Animals (Basel) ; 13(18)2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37760333

ABSTRACT

In recent years, research on fish has seen remarkable advancements, especially in aquaculture, ornamental fish industry, and biomedical studies. Immunohistochemistry has become crucial in fish research, aiding in physiological and pathological investigations. However, the use of antibodies originally developed for mammals has raised concerns about their cross-reactivity and specificity in fish. This study systematically evaluated the reactivity of commonly used antibodies for diagnostic purposes, especially in fish pathology, including pan-cytokeratin, vimentin, S-100, glial fibrillary acidic protein, and desmin in the tissue of Sparus aurata, Dicentrarchus labrax, Oncorhynchus mykiss, and Carassius auratus. Western immunoblotting was employed to assess antibody specificity. The results revealed that the pan-cytokeratin and glial fibrillary acidic protein antibodies cross-react with all tested fish species, while S-100 demonstrated specific staining in sea bream, goldfish, and rainbow trout tissues. Conversely, vimentin and desmin antibodies displayed no reactivity. In conclusion, the anti-cytokeratin clone AE1/AE3 and the polyclonal rabbit anti-glial fibrillary acidic protein antibody, which are extensively used in mammals, were validated for fish immunohistochemical studies. Regrettably, D33 anti-desmin and V9 anti-vimentin clones are unsuitable for immunohistochemistry in the tested fish. These findings underscore the need for species-specific antibodies and proper validation for accurate immunohistochemistry analyses in fish research.

2.
Animals (Basel) ; 13(9)2023 May 06.
Article in English | MEDLINE | ID: mdl-37174600

ABSTRACT

Histopathology, the gold-standard technique in classifying canine mammary tumors (CMTs), is a time-consuming process, affected by high inter-observer variability. Digital (DP) and Computer-aided pathology (CAD) are emergent fields that will improve overall classification accuracy. In this study, the ability of the CAD systems to distinguish benign from malignant CMTs has been explored on a dataset-namely CMTD-of 1056 hematoxylin and eosin JPEG images from 20 benign and 24 malignant CMTs, with three different CAD systems based on the combination of a convolutional neural network (VGG16, Inception v3, EfficientNet), which acts as a feature extractor, and a classifier (support vector machines (SVM) or stochastic gradient boosting (SGB)), placed on top of the neural net. Based on a human breast cancer dataset (i.e., BreakHis) (accuracy from 0.86 to 0.91), our models were applied to the CMT dataset, showing accuracy from 0.63 to 0.85 across all architectures. The EfficientNet framework coupled with SVM resulted in the best performances with an accuracy from 0.82 to 0.85. The encouraging results obtained by the use of DP and CAD systems in CMTs provide an interesting perspective on the integration of artificial intelligence and machine learning technologies in cancer-related research.

3.
Int J Mol Sci ; 24(1)2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36613478

ABSTRACT

The genus Anaplasma (Anaplasmataceae, Rickettsiales) includes tick-transmitted bacterial species of importance to both veterinary and human medicine. Apart from the traditionally recognized six Anaplasma species (A. phagocytophilum, A. platys, A. bovis, A. ovis, A. centrale, A. marginale), novel strains and candidate species, also of relevance to veterinary and human medicine, are emerging worldwide. Although species related to the zoonotic A. platys and A. phagocytophilum have been reported in several African and European Mediterranean countries, data on the presence of these species in sub-Saharan countries are still lacking. This manuscript reports the investigation of Anaplasma strains related to zoonotic species in ruminants in Senegal by combining different molecular tests and phylogenetic approaches. The results demonstrated a recent introduction of Candidatus (Ca) Anaplasma turritanum, a species related to the pathogenic A. platys, possibly originating by founder effect. Further, novel undetected strains related to Candidatus (Ca) Anaplasma cinensis were detected in cattle. Based on groEL and gltA molecular comparisons, we propose including these latter strains into the Candidatus (Ca) Anaplasma africanum species. Finally, we also report the emergence of Candidatus (Ca) A. boleense in Senegal. Collectively, results confirm that Anaplasma species diversity is greater than expected and should be further investigated, and that Anaplasma routine diagnostic procedures and epidemiological surveillance should take into account specificity issues raised by the presence of these novel strains, suggesting the use of a One Health approach for the management of Anaplasmataceae in sub-Saharan Africa.


Subject(s)
Anaplasma , Anaplasmataceae , Humans , Animals , Cattle , Sheep , Anaplasma/genetics , Phylogeny , Senegal/epidemiology , Anaplasmataceae/genetics , Ruminants , RNA, Ribosomal, 16S
4.
Cell Death Differ ; 28(10): 2970-2982, 2021 10.
Article in English | MEDLINE | ID: mdl-34226680

ABSTRACT

Junctional adhesion molecules (JAMs) play a critical role in cell permeability, polarity and migration. JAM-A, a key protein of the JAM family, is altered in a number of conditions including cancer; however, consequences of JAM-A dysregulation on carcinogenesis appear to be tissue dependent and organ dependent with significant implications for the use of JAM-A as a biomarker or therapeutic target. Here, we test the expression and prognostic role of JAM-A downregulation in primary and metastatic colorectal cancer (CRC) (n = 947). We show that JAM-A downregulation is observed in ~60% of CRC and correlates with poor outcome in four cohorts of stages II and III CRC (n = 1098). Using JAM-A knockdown, re-expression and rescue experiments in cell line monolayers, 3D spheroids, patient-derived organoids and xenotransplants, we demonstrate that JAM-A silencing promotes proliferation and migration in 2D and 3D cell models and increases tumour volume and metastases in vivo. Using gene-expression and proteomic analyses, we show that JAM-A downregulation results in the activation of ERK, AKT and ROCK pathways and leads to decreased bone morphogenetic protein 7 expression. We identify MIR21 upregulation as the cause of JAM-A downregulation and show that JAM-A rescue mitigates the effects of MIR21 overexpression on cancer phenotype. Our results identify a novel molecular loop involving MIR21 dysregulation, JAM-A silencing and activation of multiple oncogenic pathways in promoting invasiveness and metastasis in CRC.


Subject(s)
Cell Adhesion Molecules/metabolism , Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Receptors, Cell Surface/metabolism , Animals , Case-Control Studies , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Female , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Metastasis , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
5.
Nat Cancer ; 1(12): 1153-1166, 2020 12.
Article in English | MEDLINE | ID: mdl-33644766

ABSTRACT

Intratumoral regulatory T cell (Treg) abundance associates with diminished anti-tumor immunity and poor prognosis in human cancers. Recent work demonstrates that CD25, the high affinity receptor subunit for IL-2, is a selective target for Treg depletion in mouse and human malignancies; however, anti-human CD25 antibodies have failed to deliver clinical responses against solid tumors due to bystander IL-2 receptor signaling blockade on effector T cells, which limits their anti-tumor activity. Here we demonstrate potent single-agent activity of anti-CD25 antibodies optimized to deplete Tregs whilst preserving IL-2-STAT5 signaling on effector T cells, and demonstrate synergy with immune checkpoint blockade in vivo. Pre-clinical evaluation of an anti-human CD25 (RG6292) antibody with equivalent features demonstrates, in both non-human primates and humanized mouse models, efficient Treg depletion with no overt immune-related toxicities. Our data supports the clinical development of RG6292 and evaluation of novel combination therapies incorporating non-IL-2 blocking anti-CD25 antibodies in clinical studies.


Subject(s)
Interleukin-2 , Neoplasms , Animals , Antibodies, Monoclonal/pharmacology , Interleukin-2/pharmacology , Mice , Signal Transduction , T-Lymphocytes, Regulatory
6.
Cell Death Differ ; 24(10): 1681-1693, 2017 10.
Article in English | MEDLINE | ID: mdl-28622298

ABSTRACT

Recent studies have suggested increased plasticity of differentiated cells within the intestine to act both as intestinal stem cells (ISCs) and tumour-initiating cells. However, little is known of the processes that regulate this plasticity. Our previous work has shown that activating mutations of Kras or the NF-κB pathway can drive dedifferentiation of intestinal cells lacking Apc. To investigate this process further, we profiled both cells undergoing dedifferentiation in vitro and tumours generated from these cells in vivo by gene expression analysis. Remarkably, no clear differences were observed in the tumours; however, during dedifferentiation in vitro we found a marked upregulation of TGFß signalling, a pathway commonly mutated in colorectal cancer (CRC). Genetic inactivation of TGFß type 1 receptor (Tgfbr1/Alk5) enhanced the ability of KrasG12D/+ mutation to drive dedifferentiation and markedly accelerated tumourigenesis. Mechanistically this is associated with a marked activation of MAPK signalling. Tumourigenesis from differentiated compartments is potently inhibited by MEK inhibition. Taken together, we show that tumours arising in differentiated compartments will be exposed to different suppressive signals, for example, TGFß and blockade of these makes tumourigenesis more efficient from this compartment.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , MAP Kinase Signaling System , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/genetics , Animals , Cell Proliferation/genetics , Genes, ras/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , NF-kappa B/metabolism , Transforming Growth Factor beta/genetics
7.
Gut ; 66(7): 1268-1277, 2017 07.
Article in English | MEDLINE | ID: mdl-27618837

ABSTRACT

OBJECTIVE: Transcribed-ultraconserved regions (T-UCR) are long non-coding RNAs which are conserved across species and are involved in carcinogenesis. We studied T-UCRs downstream of the Wnt/ß-catenin pathway in liver cancer. DESIGN: Hypomorphic Apc mice (Apcfl/fl) and thiocetamide (TAA)-treated rats developed Wnt/ß-catenin dependent hepatocarcinoma (HCC) and cholangiocarcinoma (CCA), respectively. T-UCR expression was assessed by microarray, real-time PCR and in situ hybridisation. RESULTS: Overexpression of the T-UCR uc.158- could differentiate Wnt/ß-catenin dependent HCC from normal liver and from ß-catenin negative diethylnitrosamine (DEN)-induced HCC. uc.158- was overexpressed in human HepG2 versus Huh7 cells in line with activation of the Wnt pathway. In vitro modulation of ß-catenin altered uc.158- expression in human malignant hepatocytes. uc.158- expression was increased in CTNNB1-mutated human HCCs compared with non-mutated human HCCs, and in human HCC with nuclear localisation of ß-catenin. uc.158- was increased in TAA rat CCA and reduced after treatment with Wnt/ß-catenin inhibitors. uc.158- expression was negative in human normal liver and biliary epithelia, while it was increased in human CCA in two different cohorts. Locked nucleic acid-mediated inhibition of uc.158- reduced anchorage cell growth, 3D-spheroid formation and spheroid-based cell migration, and increased apoptosis in HepG2 and SW1 cells. miR-193b was predicted to have binding sites within the uc.158- sequence. Modulation of uc.158- changed miR-193b expression in human malignant hepatocytes. Co-transfection of uc.158- inhibitor and anti-miR-193b rescued the effect of uc.158- inhibition on cell viability. CONCLUSIONS: We showed that uc.158- is activated by the Wnt pathway in liver cancers and drives their growth. Thus, it may represent a promising target for the development of novel therapeutics.


Subject(s)
Bile Duct Neoplasms/metabolism , Carcinoma, Hepatocellular/metabolism , Cholangiocarcinoma/metabolism , Conserved Sequence/genetics , Liver Neoplasms/metabolism , RNA, Untranslated/genetics , Wnt Signaling Pathway , Animals , Bile Duct Neoplasms/genetics , Carcinoma, Hepatocellular/genetics , Cholangiocarcinoma/genetics , Gene Expression Regulation, Neoplastic/genetics , Hepatocytes/metabolism , Humans , Liver Neoplasms/genetics , Mice, Knockout , MicroRNAs/metabolism , Neoplasms, Experimental , Transfection , beta Catenin/genetics , beta Catenin/metabolism
8.
Nat Commun ; 7: 13615, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27897178

ABSTRACT

The multigenic nature of human tumours presents a fundamental challenge for cancer drug discovery. Here we use Drosophila to generate 32 multigenic models of colon cancer using patient data from The Cancer Genome Atlas. These models recapitulate key features of human cancer, often as emergent properties of multigenic combinations. Multigenic models such as ras p53 pten apc exhibit emergent resistance to a panel of cancer-relevant drugs. Exploring one drug in detail, we identify a mechanism of resistance for the PI3K pathway inhibitor BEZ235. We use this data to identify a combinatorial therapy that circumvents this resistance through a two-step process of emergent pathway dependence and sensitivity we term 'induced dependence'. This approach is effective in cultured human tumour cells, xenografts and mouse models of colorectal cancer. These data demonstrate how multigenic animal models that reference cancer genomes can provide an effective approach for developing novel targeted therapies.


Subject(s)
Colorectal Neoplasms/genetics , Drosophila melanogaster/genetics , Genome , Genomics , Acetates/pharmacology , Acetates/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzopyrans/pharmacology , Benzopyrans/therapeutic use , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Transformed , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epistasis, Genetic/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Reproducibility of Results , Signal Transduction/drug effects
9.
J Virol ; 89(1): 535-44, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25339764

ABSTRACT

UNLABELLED: Bone marrow stromal cell antigen 2 (BST2) is a cellular restriction factor with a broad antiviral activity. In sheep, the BST2 gene is duplicated into two paralogs termed oBST2A and oBST2B. oBST2A impedes viral exit of the Jaagsiekte sheep retroviruses (JSRV), most probably by retaining virions at the cell membrane, similar to the "tethering" mechanism exerted by human BST2. In this study, we provide evidence that unlike oBST2A, oBST2B is limited to the Golgi apparatus and disrupts JSRV envelope (Env) trafficking by sequestering it. In turn, oBST2B leads to a reduction in Env incorporation into viral particles, which ultimately results in the release of virions that are less infectious. Furthermore, the activity of oBST2B does not seem to be restricted to retroviruses, as it also acts on vesicular stomatitis virus glycoproteins. Therefore, we suggest that oBST2B exerts antiviral activity using a mechanism distinct from the classical tethering restriction observed for oBST2A. IMPORTANCE: BST2 is a powerful cellular restriction factor against a wide range of enveloped viruses. Sheep possess two paralogs of the BST2 gene called oBST2A and oBST2B. JSRV, the causative agent of a transmissible lung cancer of sheep, is known to be restricted by oBST2A. In this study, we show that unlike oBST2A, oBST2B impairs the normal cellular trafficking of JSRV envelope glycoproteins by sequestering them within the Golgi apparatus. We also show that oBST2B decreases the incorporation of envelope glycoprotein into JSRV viral particles, which in turn reduces virion infectivity. In conclusion, oBST2B exerts a novel antiviral activity that is distinct from those of BST2 proteins of other species.


Subject(s)
Jaagsiekte sheep retrovirus/immunology , Jaagsiekte sheep retrovirus/physiology , Membrane Glycoproteins/immunology , Viral Envelope Proteins/antagonists & inhibitors , Virion/metabolism , Virus Assembly , Animals , Golgi Apparatus/metabolism , Protein Transport , Sheep
10.
Cancer Cell ; 25(4): 469-83, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24735923

ABSTRACT

MicroRNA deregulation is frequent in human colorectal cancers (CRCs), but little is known as to whether it represents a bystander event or actually drives tumor progression in vivo. We show that miR-135b overexpression is triggered in mice and humans by APC loss, PTEN/PI3K pathway deregulation, and SRC overexpression and promotes tumor transformation and progression. We show that miR-135b upregulation is common in sporadic and inflammatory bowel disease-associated human CRCs and correlates with tumor stage and poor clinical outcome. Inhibition of miR-135b in CRC mouse models reduces tumor growth by controlling genes involved in proliferation, invasion, and apoptosis. We identify miR-135b as a key downsteam effector of oncogenic pathways and a potential target for CRC treatment.


Subject(s)
Colonic Neoplasms/genetics , MicroRNAs/genetics , Animals , Cell Growth Processes/genetics , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Disease Progression , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Nude , MicroRNAs/metabolism , Transfection
11.
J Virol ; 87(19): 10752-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903827

ABSTRACT

Understanding the factors governing host species barriers to virus transmission has added significantly to our appreciation of virus pathogenesis. Jaagsiekte sheep retrovirus (JSRV) is the causative agent of ovine pulmonary adenocarcinoma (OPA), a transmissible lung cancer of sheep that has rarely been found in goats. In this study, in order to further clarify the pathogenesis of OPA, we investigated whether goats are resistant to JSRV replication and carcinogenesis. We found that JSRV induces lung tumors in goats with macroscopic and histopathological features that dramatically differ from those in sheep. However, the origins of the tumor cells in the two species are identical. Interestingly, in experimentally infected lambs and goat kids, we revealed major differences in the number of virus-infected cells at early stages of infection. These differences were not related to the number of available target cells for virus infection and cell transformation or the presence of a host-specific immune response toward JSRV. Indeed, we also found that goats possess transcriptionally active endogenous retroviruses (enJSRVs) that likely influence the host immune response toward the exogenous JSRV. Overall, these results suggest that goat cells, or at least those cells targeted for viral carcinogenesis, are not permissive to virus replication but can be transformed by JSRV.


Subject(s)
Adenocarcinoma/etiology , Cell Transformation, Neoplastic/pathology , Host-Pathogen Interactions , Jaagsiekte sheep retrovirus/pathogenicity , Lung Neoplasms/etiology , Pulmonary Adenomatosis, Ovine/virology , Virus Replication , Adenocarcinoma/pathology , Animals , Blotting, Western , Cells, Cultured , Female , Fluorescent Antibody Technique , Goats , Immunoenzyme Techniques , In Situ Hybridization , Jaagsiekte sheep retrovirus/physiology , Lung Neoplasms/pathology , Pulmonary Adenomatosis, Ovine/complications , Pulmonary Adenomatosis, Ovine/pathology , RNA, Messenger/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sheep
12.
PLoS Pathog ; 9(1): e1003133, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23326235

ABSTRACT

Schmallenberg virus (SBV) is an emerging orthobunyavirus of ruminants associated with outbreaks of congenital malformations in aborted and stillborn animals. Since its discovery in November 2011, SBV has spread very rapidly to many European countries. Here, we developed molecular and serological tools, and an experimental in vivo model as a platform to study SBV pathogenesis, tropism and virus-host cell interactions. Using a synthetic biology approach, we developed a reverse genetics system for the rapid rescue and genetic manipulation of SBV. We showed that SBV has a wide tropism in cell culture and "synthetic" SBV replicates in vitro as efficiently as wild type virus. We developed an experimental mouse model to study SBV infection and showed that this virus replicates abundantly in neurons where it causes cerebral malacia and vacuolation of the cerebral cortex. These virus-induced acute lesions are useful in understanding the progression from vacuolation to porencephaly and extensive tissue destruction, often observed in aborted lambs and calves in naturally occurring Schmallenberg cases. Indeed, we detected high levels of SBV antigens in the neurons of the gray matter of brain and spinal cord of naturally affected lambs and calves, suggesting that muscular hypoplasia observed in SBV-infected lambs is mostly secondary to central nervous system damage. Finally, we investigated the molecular determinants of SBV virulence. Interestingly, we found a biological SBV clone that after passage in cell culture displays increased virulence in mice. We also found that a SBV deletion mutant of the non-structural NSs protein (SBVΔNSs) is less virulent in mice than wild type SBV. Attenuation of SBV virulence depends on the inability of SBVΔNSs to block IFN synthesis in virus infected cells. In conclusion, this work provides a useful experimental framework to study the biology and pathogenesis of SBV.


Subject(s)
Bunyaviridae Infections/virology , Cerebral Cortex/virology , Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Orthobunyavirus/pathogenicity , Amino Acid Sequence , Animals , Base Sequence , Bunyaviridae Infections/immunology , Bunyaviridae Infections/mortality , Bunyaviridae Infections/pathology , Cattle , Cell Line , Cerebellar Diseases/immunology , Cerebellar Diseases/pathology , Cerebellar Diseases/virology , Cerebral Cortex/immunology , Cerebral Cortex/pathology , Disease Models, Animal , Disease Progression , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Mice , Molecular Sequence Data , Neurons/immunology , Neurons/pathology , Neurons/virology , Orthobunyavirus/genetics , Orthobunyavirus/isolation & purification , Sequence Deletion , Sheep , Spinal Cord/immunology , Spinal Cord/pathology , Spinal Cord/virology , Survival Rate , Vacuoles , Viral Tropism , Virulence , Virus Cultivation , Virus Replication
13.
J Virol ; 85(14): 7118-28, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21593182

ABSTRACT

The exogenous and pathogenic Jaagsiekte sheep retrovirus (JSRV) coexists with highly related and biologically active endogenous retroviruses (enJSRVs). The endogenous enJS56A1 locus possesses a defective Gag polyprotein which blocks the late replication steps of related exogenous and endogenous retroviruses by a mechanism known as JSRV late restriction (JLR). Conversely, enJSRV-26, which most likely integrated into the sheep genome less than 200 years ago, is able to escape JLR. In this study, we demonstrate that the ability of enJSRV-26 to escape JLR is due to a single-amino-acid substitution in the signal peptide (SP) of its envelope glycoprotein. We show that enJSRV-26 SP does not localize to the nucleolus, unlike the functional SPs of related exogenous and endogenous sheep betaretroviruses. In addition, enJSRV-26 SP function as a posttranscriptional regulator of viral gene expression is impaired. enJSRV-26 JLR escape relies on the presence of the functional enJS56A1 SP. Moreover, we show that the ratio between enJSRV-26 and enJS56A1 Gag is critical to elude JLR. Interestingly, we found that the domestic sheep has acquired, by genome amplification, several copies of the enJS56A1 provirus. These data further reinforce the notion that transdominant enJSRV proviruses have been positively selected in domestic sheep, and that the coevolution between endogenous and exogenous sheep betaretroviruses and their host is still occurring.


Subject(s)
Betaretrovirus/physiology , Genes, gag , Protein Sorting Signals , Animals , Betaretrovirus/metabolism , Blotting, Western , COS Cells , Cell Line , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Humans , Microscopy, Confocal , Polymerase Chain Reaction , Sheep
14.
PLoS Pathog ; 7(3): e1002014, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21483485

ABSTRACT

Jaagsiekte sheep retrovirus (JSRV) is a unique oncogenic virus with distinctive biological properties. JSRV is the only virus causing a naturally occurring lung cancer (ovine pulmonary adenocarcinoma, OPA) and possessing a major structural protein that functions as a dominant oncoprotein. Lung cancer is the major cause of death among cancer patients. OPA can be an extremely useful animal model in order to identify the cells originating lung adenocarcinoma and to study the early events of pulmonary carcinogenesis. In this study, we demonstrated that lung adenocarcinoma in sheep originates from infection and transformation of proliferating type 2 pneumocytes (termed here lung alveolar proliferating cells, LAPCs). We excluded that OPA originates from a bronchioalveolar stem cell, or from mature post-mitotic type 2 pneumocytes or from either proliferating or non-proliferating Clara cells. We show that young animals possess abundant LAPCs and are highly susceptible to JSRV infection and transformation. On the contrary, healthy adult sheep, which are normally resistant to experimental OPA induction, exhibit a relatively low number of LAPCs and are resistant to JSRV infection of the respiratory epithelium. Importantly, induction of lung injury increased dramatically the number of LAPCs in adult sheep and rendered these animals fully susceptible to JSRV infection and transformation. Furthermore, we show that JSRV preferentially infects actively dividing cell in vitro. Overall, our study provides unique insights into pulmonary biology and carcinogenesis and suggests that JSRV and its host have reached an evolutionary equilibrium in which productive infection (and transformation) can occur only in cells that are scarce for most of the lifespan of the sheep. Our data also indicate that, at least in this model, inflammation can predispose to retroviral infection and cancer.


Subject(s)
Adenocarcinoma/veterinary , Alveolar Epithelial Cells/virology , Cell Transformation, Viral , Jaagsiekte sheep retrovirus/pathogenicity , Lung Neoplasms/veterinary , Pulmonary Adenomatosis, Ovine/pathology , Pulmonary Adenomatosis, Ovine/virology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Humans , Inflammation/immunology , Lung/embryology , Lung Neoplasms/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Sheep , Viral Structural Proteins/metabolism
15.
J Virol ; 83(9): 4591-604, 2009 May.
Article in English | MEDLINE | ID: mdl-19244321

ABSTRACT

Retroviruses use different strategies to regulate transcription and translation and exploit the cellular machinery involved in these processes. This study shows that the signal peptide of the envelope glycoprotein (Env) of Jaagsiekte sheep retrovirus (JSRV) plays a major role in posttranscriptional viral gene expression. Expression of the JSRV Env in trans increases viral particle production by mechanisms dependent on (i) its leader sequence, (ii) an intact signal peptide cleavage site, (iii) a cis-acting RNA-responsive element located in the viral genome, (iv) Crm1, and (v) B23. The signal peptide of the JSRV Env (JSE-SP) is 80 amino acid residues in length and contains putative nuclear localization and export signals, in addition to an arginine-rich RNA binding motif. JSE-SP localizes both in the endoplasmic reticulum and in the nucleus, where it colocalizes with nucleolar markers. JSE-SP is a multifunctional protein, as it moderately enhances nuclear export of unspliced viral mRNA and considerably increases viral particle release by favoring a posttranslational step of the replication cycle.


Subject(s)
Gene Expression Regulation, Viral/genetics , Protein Processing, Post-Translational/genetics , Protein Sorting Signals/genetics , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleolus/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Genome, Viral/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Jaagsiekte sheep retrovirus/genetics , Jaagsiekte sheep retrovirus/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Protein Transport , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription, Genetic/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Virion/metabolism , Exportin 1 Protein
16.
Virology ; 367(2): 413-21, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17610928

ABSTRACT

Jaagsiekte retrovirus (JSRV) causes ovine pulmonary adenocarcinoma (OPA), a transmissible lung cancer of sheep. The envelope (Env) glycoprotein protein of JSRV functions as a dominant oncoprotein in vitro and in vivo. An SH2 binding domain (YXXM) in the cytoplasmic tail of the JSRV Env is one of the main determinants of viral transformation at least in vitro. In these studies, we report the first in vivo tests of site-specific mutants of JSRV in their natural host, the sheep. We show that, in vivo, JSRV(21) with the cytoplasmic tail YXXM mutated to DXXM did not cause disease nor detectable infection, indicating that this motif is absolutely required for virus replication and possibly transformation in vivo. In contrast, mutation of the JSRV open reading frame orfX, for which no function has yet been attributed, did not alter the disease induced by JSRV(21).


Subject(s)
Cell Transformation, Viral/physiology , Jaagsiekte sheep retrovirus/genetics , Jaagsiekte sheep retrovirus/physiology , Viral Envelope Proteins/physiology , Amino Acid Sequence , Animals , Carcinogenicity Tests , Cell Line , Cytoplasm/chemistry , Jaagsiekte sheep retrovirus/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Open Reading Frames , Sheep , Viral Envelope Proteins/chemistry
17.
Cell ; 126(3): 477-87, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16901782

ABSTRACT

The transmissible agent causing canine transmissible venereal tumor (CTVT) is thought to be the tumor cell itself. To test this hypothesis, we analyzed genetic markers including major histocompatibility (MHC) genes, microsatellites, and mitochondrial DNA (mtDNA) in naturally occurring tumors and matched blood samples. In each case, the tumor is genetically distinct from its host. Moreover, tumors collected from 40 dogs in 5 continents are derived from a single neoplastic clone that has diverged into two subclades. Phylogenetic analyses indicate that CTVT most likely originated from a wolf or an East Asian breed of dog between 200 and 2500 years ago. Although CTVT is highly aneuploid, it has a remarkably stable genotype. During progressive growth, CTVT downmodulates MHC antigen expression. Our findings have implications for understanding genome instability in cancer, natural transplantation of allografts, and the capacity of a somatic cell to evolve into a transmissible parasite.


Subject(s)
Canidae/genetics , Cell Lineage/genetics , Dog Diseases/genetics , Dog Diseases/transmission , Evolution, Molecular , Venereal Tumors, Veterinary/genetics , Aneuploidy , Animals , Base Sequence , Clone Cells/cytology , Clone Cells/metabolism , DNA, Mitochondrial/genetics , Dogs , Down-Regulation/genetics , Female , Genetic Markers , Genomic Instability/genetics , Genotype , Histocompatibility Antigens/genetics , Immune Tolerance/genetics , Male , Microsatellite Repeats/genetics , Molecular Sequence Data , Phylogeny , Sexual Behavior, Animal
18.
J Virol ; 76(11): 5387-94, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11991967

ABSTRACT

Ovine betaretroviruses include Jaagsiekte sheep retrovirus (JSRV) and enzootic nasal tumor virus (ENTV). JSRV and ENTV represent a unique class of oncogenic retroviruses that induce tumors of the respiratory tract. JSRV and ENTV are highly related but induce different diseases. Expression of the JSRV envelope (Env) induces transformation of rodent fibroblasts in vitro and phosphorylation of Akt, a central player in the phosphatidylinositol 3-kinase (PI-3K)/Akt signal transduction pathway. However, little information is available on the molecular biology of ENTV. In this study, we initially assessed whether the ENTV Env has the same properties as the homologous JSRV protein. We performed entry and interference assays using retroviral vectors pseudotyped with either the JSRV or the ENTV Env and sheep choroid plexus cells, choroid plexus cells stably expressing the JSRV Env protein, human 293T cells, mouse NIH 3T3 cells, or NIH 3T3 cells expressing human hyaluronidase 2 (HYAL2), the cellular receptor for JSRV. The results obtained indicated that ENTV and JSRV share the same receptor in sheep cells and that they can use human HYAL2 as a cellular receptor in mouse cells. The ENTV Env induces transformation of rodent fibroblasts in vitro. As with the JSRV Env, the tyrosine at position 590 is critical for ENTV Env-induced cell transformation, and Akt is phosphorylated in ENTV Env-transformed cells but not in the parental cell lines. Thus, ovine betaretroviruses share a common mechanism of cell transformation. We further investigated the relevance of Akt activation in cells transformed by ovine betaretroviruses. A PI-3K inhibitor blocked Akt phosphorylation in JSRV Env-transformed cells, suggesting a possible involvement of PI-3K in JSRV and ENTV Env-induced cell transformation. In addition, phosphorylated Akt was detected in a cell line derived from a lung tumor of a sheep with naturally occurring ovine pulmonary adenocarcinoma.


Subject(s)
Betaretrovirus/genetics , Cell Transformation, Viral , Gene Products, env/genetics , Jaagsiekte sheep retrovirus/genetics , Protein Serine-Threonine Kinases , 3T3 Cells , Amino Acid Sequence , Animals , Betaretrovirus/physiology , Cell Line , Cell Line, Transformed , Cytoplasm/metabolism , Gene Products, env/metabolism , Gene Products, env/physiology , Humans , Jaagsiekte sheep retrovirus/physiology , Mice , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Receptors, Virus/metabolism , Sheep , Tumor Cells, Cultured
19.
Virology ; 294(1): 180-8, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11886276

ABSTRACT

Jaagsiekte sheep retrovirus (JSRV) is the etiologic agent of a contagious lung cancer of sheep, ovine pulmonary adenocarcinoma (OPA). In this study, we characterized the virus-specific RNAs in 293T cells transiently transfected with a human cytomegalovirus promoter-driven JSRV expression plasmid, in productively infected OHH1.LU deer lung cells, and in OPA tumors from field isolates. Typical unspliced (presumably for gag, pro, and pol) and singly spliced env mRNAs were detected. In addition, six other virus-specific RNAs were detected that resulted from the use of alternate splice acceptor sites and two premature polyadenylation sites (located in gag and in env). The orf-x gene of the virus appears to be expressed from two singly spliced subgenomic mRNAs of 3.2 kb that would encode an independent orf-x protein of 179 amino acids. In addition, the results suggested that there may also be an internal promoter for orf-x.


Subject(s)
Jaagsiekte sheep retrovirus/pathogenicity , Pulmonary Adenomatosis, Ovine/virology , RNA Splicing , RNA, Messenger/metabolism , RNA, Viral/metabolism , Transfection , Animals , Base Sequence , Cell Line , Jaagsiekte sheep retrovirus/genetics , Molecular Sequence Data , Polyadenylation , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...