Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.073
Filter
1.
Proc Natl Acad Sci U S A ; 120(51): e2316467120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38079542

ABSTRACT

Merkel cell polyomavirus (MCV or MCPyV) is an alphapolyomavirus causing human Merkel cell carcinoma and encodes four tumor (T) antigen proteins: large T (LT), small tumor (sT), 57 kT, and middle T (MT)/alternate LT open reading frame proteins. We show that MCV MT is generated as multiple isoforms through internal methionine translational initiation that insert into membrane lipid rafts. The membrane-localized MCV MT oligomerizes and promiscuously binds to lipid raft-associated Src family kinases (SFKs). MCV MT-SFK interaction is mediated by a Src homology (SH) 3 recognition motif as determined by surface plasmon resonance, coimmunoprecipitation, and bimolecular fluorescence complementation assays. SFK recruitment by MT leads to tyrosine phosphorylation at a SH2 recognition motif (pMTY114), allowing interaction with phospholipase C gamma 1 (PLCγ1). The secondary recruitment of PLCγ1 to the SFK-MT membrane complex promotes PLCγ1 tyrosine phosphorylation on Y783 and activates the NF-κB inflammatory signaling pathway. Mutations at either the MCV MT SH2 or SH3 recognition sites abrogate PLCγ1-dependent activation of NF-κB signaling and increase viral replication after MCV genome transfection into 293 cells. These findings reveal a conserved viral targeting of the SFK-PLCγ1 pathway by both MCV and murine polyomavirus (MuPyV) MT proteins. The molecular steps in how SFK-PLCγ1 activation is achieved, however, differ between these two viruses.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Mice , Animals , Humans , Antigens, Polyomavirus Transforming/metabolism , Merkel cell polyomavirus/metabolism , NF-kappa B/metabolism , src-Family Kinases/metabolism , Phospholipase C gamma/metabolism , Signal Transduction , Antigens, Viral, Tumor/genetics , Carcinoma, Merkel Cell/genetics , Tyrosine/metabolism
2.
Proc Natl Acad Sci U S A ; 120(30): e2308010120, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37459531

ABSTRACT

Cellular eukaryotic replication initiation helicases are first loaded as head-to-head double hexamers on double-stranded (ds) DNA origins and then initiate S-phase DNA melting during licensed (once per cell cycle) replication. Merkel cell polyomavirus (MCV) large T (LT) helicase oncoprotein similarly binds and melts its own 98-bp origin but replicates multiple times in a single cell cycle. To examine the actions of this unlicensed viral helicase, we quantitated multimerization of MCV LT molecules as they assembled on MCV DNA origins using real-time single-molecule microscopy. MCV LT formed highly stable double hexamers having 17-fold longer mean lifetime (τ, >1,500 s) on DNA than single hexamers. Unexpectedly, partial MCV LT assembly without double-hexamer formation was sufficient to melt origin dsDNA as measured by RAD51, RPA70, or S1 nuclease cobinding. DNA melting also occurred with truncated MCV LT proteins lacking the helicase domain, but was lost from a protein without the multimerization domain that could bind only as a monomer to DNA. SV40 polyomavirus LT also multimerized to the MCV origin without forming a functional hexamer but still melted origin DNA. MCV origin melting did not require ATP hydrolysis and occurred for both MCV and SV40 LT proteins using the nonhydrolyzable ATP analog, adenylyl-imidodiphosphate (AMP-PNP). LT double hexamers formed in AMP-PNP, and melted DNA, consistent with direct LT hexamer assembly around single-stranded (ss) DNA without the energy-dependent dsDNA-to-ssDNA melting and remodeling steps used by cellular helicases. These results indicate that LT multimerization rather than helicase activity is required for origin DNA melting during unlicensed virus replication.


Subject(s)
Antigens, Polyomavirus Transforming , Simian virus 40 , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Simian virus 40/genetics , Simian virus 40/metabolism , Nucleic Acid Denaturation , Adenylyl Imidodiphosphate , DNA Replication , DNA/genetics , DNA/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA, Single-Stranded , DNA, Viral/genetics , DNA, Viral/metabolism
3.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614338

ABSTRACT

Merkel cell polyomavirus (MCPyV) is the major cause of Merkel cell carcinoma (MCC), an aggressive skin cancer. MCPyV large T-antigen (LTag) and small T-antigen (sTag) are the main oncoproteins involved in MCPyV-induced MCC. A hallmark of MCPyV-positive MCC cells is the expression of a C-terminal truncated LTag. Protein kinase A (PKA) plays a fundamental role in a variety of biological processes, including transcription by phosphorylating and thereby regulating the activity of transcription factors. As MCPyV LTag has been shown to be phosphorylated and acts as a transcription factor for the viral early and late promoter, we investigated whether LTag can be phosphorylayted by PKA, and whether this affects the transcript activity of LTag. Using a phosphorylation prediction algorithm, serine 191, 203, and 265 were identified as putative phosphorylation sites for PKA. Mass spectrometry of in vitro PKA-phosphorylated peptides confirmed phosphorylation of S203 and S265, but not S191. Full-length LTag inhibited early and late promoter activity of MCPyV, whereas the truncated MKL2 LTag variant stimulated both promoters. Single non-phosphorylable, as well as phosphomimicking mutations did not alter the inhibitory effect of full-length LTag. However, the non-phosphorylable mutations abrogated transactivation of the MCPyV promoters by MKL2 LTag, whereas phosphomimicking substitutions restored the ability of MKL2 LTag to activate the promoters. Triple LTag and MKL2 LTag mutants had the same effect as the single mutants. Activation of the PKA signaling pathway did not enhance MCPyV promoter activity, nor did it affect LTag expression levels in MCPyV-positive Merkel cell carcinoma (MCC) cells. Our results show that phosphorylation of truncated LTag stimulates viral promoter activity, which may contribute to higher levels of the viral oncoproteins LTag and sTag. Interfering with PKA-induced LTag phosphorylation/activity may be a therapeutic strategy to treat MCPyV-positive MCC patients.


Subject(s)
Antigens, Polyomavirus Transforming , Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Tumor Virus Infections , Humans , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/virology , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Merkel cell polyomavirus/metabolism , Phosphorylation , Polyomavirus Infections/metabolism , Polyomavirus Infections/virology , Skin Neoplasms/metabolism , Skin Neoplasms/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Antigens, Polyomavirus Transforming/metabolism , Transcription, Genetic
4.
PLoS Pathog ; 18(5): e1010551, 2022 05.
Article in English | MEDLINE | ID: mdl-35560034

ABSTRACT

Clear evidence supports a causal link between Merkel cell polyomavirus (MCPyV) and the highly aggressive human skin cancer called Merkel cell carcinoma (MCC). Integration of viral DNA into the human genome facilitates continued expression of the MCPyV small tumor (ST) and large tumor (LT) antigens in virus-positive MCCs. In MCC tumors, MCPyV LT is truncated in a manner that renders the virus unable to replicate yet preserves the LXCXE motif that facilitates its binding to and inactivation of the retinoblastoma tumor suppressor protein (pRb). We previously developed a MCPyV transgenic mouse model in which MCC tumor-derived ST and truncated LT expression were targeted to the stratified epithelium of the skin, causing epithelial hyperplasia, increased proliferation, and spontaneous tumorigenesis. We sought to determine if any of these phenotypes required the association between the truncated MCPyV LT and pRb. Mice were generated in which K14-driven MCPyV ST/LT were expressed in the context of a homozygous RbΔLXCXE knock-in allele that attenuates LT-pRb interactions through LT's LXCXE motif. We found that many of the phenotypes including tumorigenesis that develop in the K14-driven MCPyV transgenic mice were dependent upon LT's LXCXE-dependent interaction with pRb. These findings highlight the importance of the MCPyV LT-pRb interaction in an in vivo model for MCPyV-induced tumorigenesis.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Tumor Virus Infections , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Cell Transformation, Neoplastic , Hyperplasia/pathology , Merkel Cells/metabolism , Merkel Cells/pathology , Merkel cell polyomavirus/genetics , Mice , Skin Neoplasms/pathology
5.
Int J Mol Sci ; 22(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34576139

ABSTRACT

Basement membrane (BM) zone-associated collagen XV (ColXV) has been shown to suppress the malignancy of tumour cells, and its restin domain can inhibit angiogenesis. In human breast cancer, as well as in many other human carcinomas, ColXV is lost from the epithelial BM zone prior to tumour invasion. Here, we addressed the roles of ColXV in breast carcinogenesis using the transgenic MMTV-PyMT mouse mammary carcinoma model. We show here for the first time that the inactivation of Col15a1 in mice leads to changes in the fibrillar tumour matrix and to increased mammary tumour growth. ColXV is expressed by myoepithelial and endothelial cells in mammary tumours and is lost from the ductal BM along with the loss of the myoepithelial layer during cancer progression while persisting in blood vessels and capillaries, even in invasive tumours. However, despite the absence of anti-angiogenic restin domain, neovascularisation was reduced rather than increased in the ColXV-deficient mammary tumours compared to controls. We also show that, in robust tumour cell transplantation models or in a chemical-induced fibrosarcoma model, the inactivation of Col15a1 does not affect tumour growth or angiogenesis. In conclusion, our results support the proposed tumour suppressor function of ColXV in mammary carcinogenesis and reveal diverse roles of this collagen in different cancer types.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Collagen/deficiency , Extracellular Matrix/metabolism , Gene Deletion , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/pathology , Cell Proliferation , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Female , Fibrosarcoma/pathology , Fibrosis , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/ultrastructure , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stromal Cells/pathology , Stromal Cells/ultrastructure , Survival Analysis
6.
Cells ; 10(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34440833

ABSTRACT

RATIONALE: In recent decades, the great potential of human epicardium-derived cells (EPDCs) as an endogenous cell source for cardiac regeneration has been recognized. The limited availability and low proliferation capacity of primary human EPDCs and phenotypic differences between EPDCs obtained from different individuals hampers their reproducible use for experimental studies. AIM: To generate and characterize inducible proliferative adult human EPDCs for use in fundamental and applied research. METHODS AND RESULTS: Inducible proliferation of human EPDCs was achieved by doxycycline-controlled expression of simian virus 40 large T antigen (LT) with a repressor-based lentiviral Tet-On system. In the presence of doxycycline, these inducible EPDCs (iEPDCs) displayed high and long-term proliferation capacity. After doxycycline removal, LT expression ceased and the iEPDCs regained their cuboidal epithelial morphology. Similar to primary EPDCs, iEPDCs underwent an epithelial-to-mesenchymal transition (EMT) after stimulation with transforming growth factor ß3. This was confirmed by reverse transcription-quantitative polymerase chain reaction analysis of epithelial and mesenchymal marker gene expression and (immuno) cytochemical staining. Collagen gel-based cell invasion assays demonstrated that mesenchymal iEPDCs, like primary EPDCs, possess increased invasion and migration capacities as compared to their epithelial counterparts. Mesenchymal iEPDCs co-cultured with sympathetic ganglia stimulated neurite outgrowth similarly to primary EPDCs. CONCLUSION: Using an inducible LT expression system, inducible proliferative adult human EPDCs were generated displaying high proliferative capacity in the presence of doxycycline. These iEPDCs maintain essential epicardial characteristics with respect to morphology, EMT ability, and paracrine signaling following doxycycline removal. This renders iEPDCs a highly useful new in vitro model for studying human epicardial properties.


Subject(s)
Pericardium/metabolism , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Doxycycline/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Models, Biological , Neurites/physiology , Paracrine Communication/drug effects , Pericardium/cytology , Transforming Growth Factor beta3/pharmacology
7.
Cells ; 10(4)2021 04 19.
Article in English | MEDLINE | ID: mdl-33921698

ABSTRACT

MRCKα is a ubiquitously expressed serine/threonine kinase involved in cell contraction and F-actin turnover, which is highly amplified in human breast cancer and part of a gene expression signature for bad prognosis. Nothing is known about the in vivo function of MRCKα. To explore MRCKα function in development and in breast cancer, we generated mice lacking a functional MRCKα gene. Mice were born close to the Mendelian ratio and showed no obvious phenotype including a normal mammary gland formation. Assessing breast cancer development using the transgenic MMTV-PyMT mouse model, loss of MRCKα did not affect tumor onset, tumor growth and metastasis formation. Deleting MRCKα and its related family member MRCKß in two triple-negative breast cancer cell lines resulted in reduced invasion of MDA-MB-231 cells, but did not affect migration of 4T1 cells. Further genomic analysis of human breast cancers revealed that MRCKα is frequently co-amplified with the oncogenes ARID4B and AKT3 which might contribute to the prognostic value of MRCKα expression. Collectively, these data suggest that MRCKα might be a prognostic marker for breast cancer, but probably of limited functional importance.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Carcinogenesis/pathology , Mammary Neoplasms, Animal/metabolism , Mammary Tumor Virus, Mouse/physiology , Myotonin-Protein Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Antigens, Neoplasm/metabolism , Base Sequence , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Collagen/pharmacology , Disease Models, Animal , Female , Gels/pharmacology , Humans , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mammary Tumor Virus, Mouse/drug effects , Mice , Mice, Knockout , Mutation/genetics , Myosins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/metabolism , Phenotype , Phosphorylation/drug effects , Polymerization/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Triple Negative Breast Neoplasms/pathology
8.
Oncogene ; 40(3): 475-491, 2021 01.
Article in English | MEDLINE | ID: mdl-33235291

ABSTRACT

Breast cancer is associated with the second highest cancer-associated deaths worldwide. Therefore, understanding the key events that determine breast cancer progression, modulation of the tumor-microenvironment and metastasis, which is the main cause of cancer-associated death, are of great importance. The mammary specific polyomavirus middle T antigen overexpression mouse model (MMTV-PyMT), first published in 1992, is the most commonly used genetically engineered mouse model (GEMM) for cancer research. Mammary lesions arising in MMTV-PyMT mice follow similar molecular and histological progression as human breast tumors, making it an invaluable tool for cancer researchers and instrumental in understanding tumor biology. In this review, we will highlight key studies that demonstrate the utility of PyMT derived GEMMs in understanding the molecular basis of breast cancer progression, metastasis and highlight its use as a pre-clinical tool for therapeutic discovery.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic
9.
Viruses ; 12(10)2020 10 20.
Article in English | MEDLINE | ID: mdl-33092197

ABSTRACT

The oncogenic potential of both the polyomavirus large (LT-Ag) and small (Sm t-Ag) tumor antigens has been previously demonstrated in both tissue culture and animal models. Even the contribution of the MCPyV tumor antigens to the development of an aggressive human skin cancer, Merkel cell carcinoma, has been recently established. To date, the known primary targets of these tumor antigens include several tumor suppressors such as pRb, p53, and PP2A. However, a comprehensive list of the host proteins targeted by these proteins remains largely unknown. Here, we report the first interactome of JCV LT-Ag and Sm t-Ag by employing two independent "affinity purification/mass spectroscopy" (AP/MS) assays. The proteomics data identified novel targets for both tumor antigens while confirming some of the previously reported interactions. LT-Ag was found to primarily target the protein complexes with ATPase (v-ATPase and Smc5/6 complex), phosphatase (PP4 and PP1), and ligase (E3-ubiquitin) activities. In contrast, the major targets of Sm t-Ag were identified as Smarca1/6, AIFM1, SdhA/B, PP2A, and p53. The interactions between "LT-Ag and SdhB", "Sm t-Ag and Smarca5", and "Sm t-Ag and SDH" were further validated by biochemical assays. Interestingly, perturbations in some of the LT-Ag and Sm t-Ag targets identified in this study were previously shown to be associated with oncogenesis, suggesting new roles for both tumor antigens in novel oncogenic pathways. This comprehensive data establishes new foundations to further unravel the new roles for JCV tumor antigens in oncogenesis and the viral life cycle.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , JC Virus/metabolism , Multiprotein Complexes/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Carcinogenesis/metabolism , Chromatin/metabolism , Chromatography, Affinity , Humans , Ligases/metabolism , Mass Spectrometry , Phosphoric Monoester Hydrolases/metabolism , Polyomavirus Infections , Protein Interaction Maps , Proteomics , Tumor Virus Infections/virology , Ubiquitins/metabolism , Virus Replication
10.
Nucleic Acids Res ; 48(7): 3657-3677, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32128579

ABSTRACT

DNA replication is a central process in all living organisms. Polyomavirus DNA replication serves as a model system for eukaryotic DNA replication and has considerably contributed to our understanding of basic replication mechanisms. However, the details of the involved processes are still unclear, in particular regarding lagging strand synthesis. To delineate the complex mechanism of coordination of various cellular proteins binding simultaneously or consecutively to DNA to initiate replication, we investigated single-stranded DNA (ssDNA) interactions by the SV40 large T antigen (Tag). Using single molecule imaging by atomic force microscopy (AFM) combined with biochemical and spectroscopic analyses we reveal independent activity of monomeric and oligomeric Tag in high affinity binding to ssDNA. Depending on ssDNA length, we obtain dissociation constants for Tag-ssDNA interactions (KD values of 10-30 nM) that are in the same order of magnitude as ssDNA binding by human replication protein A (RPA). Furthermore, we observe the formation of RPA-Tag-ssDNA complexes containing hexameric as well as monomeric Tag forms. Importantly, our data clearly show stimulation of primase function in lagging strand Okazaki fragment synthesis by monomeric Tag whereas hexameric Tag inhibits the reaction, redefining DNA replication initiation on the lagging strand.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , DNA Replication , DNA, Single-Stranded/metabolism , Replication Protein A/metabolism , Adenosine Triphosphate/metabolism , DNA/metabolism , DNA Polymerase I/metabolism , DNA Primase/metabolism , DNA, Single-Stranded/chemistry , Protein Binding , Simian virus 40/immunology
11.
PLoS One ; 15(1): e0227174, 2020.
Article in English | MEDLINE | ID: mdl-31900469

ABSTRACT

BEAS-2B was originally established as an immortalized but non-tumorigenic epithelial cell line from human bronchial epithelium. Because of general recognition for its bronchial epithelial origin, the BEAS-2B cell line has been widely used as an in vitro cell model in a large variety of studies associated with respiratory diseases including lung carcinogenesis. However, very few studies have discussed non-epithelial features of BEAS-2B cells, especially the features associated with mesenchymal stem cells (MSCs), which represent a group of fibroblast-like cells with limited self-renewal and differentiation potential to various cell lineages. In this study, we compared BEAS-2B with a human umbilical cord-derived MSCs (hMSCs) cell line, hMSC1, which served as a representative of hMSCs in terms of expressing common features of hMSCs. It was observed that both BEAS-2B and hMSC1 shared the same expression profile of surface markers of hMSCs and exhibited similar osteogenic and adipogenic differentiation potential. In addition, like hMSC1, the BEAS-2B cell line exhibited suppressive activities on proliferation of mitogen-activated total T lymphocytes as well as Th1 lymphocytes, and IFNγ-induced expression of IDO1, all thus demonstrating that BEAS-2B cells exhibited an almost identical characteristic profile with hMSCs, even though, there was a clear difference between BEAS-2B and hMSCs in the effects on type 2 macrophage polarization. Most importantly, the hMSCs features of BEAS-2B were unlikely a consequence of epithelial-mesenchymal transition. Therefore, this study provided a set of evidence to provoke reconsideration of epithelial origin of BEAS-2B.


Subject(s)
Bronchi/cytology , Epithelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/cytology , A549 Cells , Animals , Antigens, Polyomavirus Transforming/metabolism , Antigens, Surface/metabolism , Carcinogenesis/metabolism , Cell Differentiation , Cell Polarity/physiology , Cell Proliferation , Coculture Techniques , Epithelial-Mesenchymal Transition , Female , Heterografts , Humans , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Th1 Cells/metabolism
12.
Appl Immunohistochem Mol Morphol ; 28(6): 444-447, 2020 07.
Article in English | MEDLINE | ID: mdl-31205069

ABSTRACT

BACKGROUND: Simian virus 40 (SV40)-contaminated polio vaccine was accidentally administered to about one-third of the UK population receiving polio vaccines between 1956 and 1962. SV40 was subsequently demonstrated to be a carcinogenic virus in experimental and animal models. Since then, the SV40 oncogenic protein large T antigen (SV40 Tag) has been shown to cause malignant transformation of asbestos-treated human pleural mesothelial cells and malignant pleural mesotheliomas in asbestos-exposed SV40 Tag transgenic mice. The present study was designed to investigate the possible association of SV40 Tag with human malignant pleural mesothelioma samples from birth cohorts of the UK population exposed to combined peak levels of asbestos and SV40-contaminated polio vaccines. MATERIALS AND METHODS: Tumor and background lung tissue microarrays prepared from archival surgical specimens of 139 pleural mesothelioma cases, collected over a period of 8 years (1998 to 2005), were analyzed. These represented birth cohorts overlapping with the period 1950 to 1960, exposed to a high level of both asbestos and SV40-contaminated live polio vaccines. SV40 Tag mRNA expression was investigated using a highly sensitive and specific SV40 Tag RNA in situ hybridization detection method on the basis of the novel RNAscope technology. RESULTS: SV40 Tag RNA was not detected in any of the 127 evaluable tumor cases, despite appropriate results obtained for the external positive and negative controls included. CONCLUSION: The complete absence of SV40 Tag mRNA in this large series of cases contradicts experimental evidence suggestive of SV40 link with asbestos-exposed malignant pleural mesotheliomas in the UK population. Alternative explanations of the negative findings are discussed to exclude possible confounding factors.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Asbestos/adverse effects , Mesothelioma, Malignant/metabolism , Pleural Neoplasms/metabolism , Poliovirus Vaccines/adverse effects , Simian virus 40/metabolism , Adult , Aged , Aged, 80 and over , Antigens, Polyomavirus Transforming/genetics , Cell Transformation, Neoplastic/genetics , Correlation of Data , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mesothelioma, Malignant/etiology , Mesothelioma, Malignant/genetics , Middle Aged , Pleural Neoplasms/etiology , Pleural Neoplasms/genetics , Pleural Neoplasms/pathology , Retrospective Studies , Simian virus 40/genetics , United Kingdom
13.
Mol Oncol ; 14(1): 211-224, 2020 01.
Article in English | MEDLINE | ID: mdl-31725949

ABSTRACT

Interleukin (IL)-22 is recognized as a tumor-supporting cytokine and is implicated in the proliferation of multiple epithelial cancers. In breast cancer, the current knowledge of IL-22 function is based on cell line models and little is known about how IL-22 affects the tumor initiation, proliferation, invasion, and metastasis in the in vivo system. Here, we investigated the tumor stage-specific function of IL-22 in disease development by evaluating the stage-by-stage progression of breast cancer in an IL-22 knockout spontaneous breast cancer mouse model. We found that among all the stages, IL-22 is specifically upregulated in tumor microenvironment (TME) during the malignant transformation stage of breast tumor progression. The deletion of IL-22 gene leads to the arrest of malignant transition stage, and reduced invasion and tumor burden. Administration of recombinant IL-22 in the TME does not influence in vivo tumor initiation and proliferation but only promotes malignant transformation of cancer cells. Mechanistically, deletion of IL-22 gene causes downregulation of epithelial-to-mesenchymal transition (EMT)-associated transcription factors in breast tumors, suggesting EMT as the mechanism of regulation of malignancy by IL-22. Clinically, in human breast tumor tissues, increased number of IL-22+ cells in the TME is associated with an aggressive phenotype of breast cancer. For the first time, this study provides an insight into the tumor stage-specific function of IL-22 in breast tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Interleukins/metabolism , Mammary Neoplasms, Experimental/metabolism , Tumor Microenvironment/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , Interleukins/administration & dosage , Interleukins/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasm Staging , Recombinant Proteins , Tissue Array Analysis , Up-Regulation
14.
J Nutr Biochem ; 77: 108313, 2020 03.
Article in English | MEDLINE | ID: mdl-31837540

ABSTRACT

Monocyte chemotactic protein-1 (MCP-1) is an adipokine with demonstrated carcinogenic potential. However, there is a lack of evidence whether adipose-produced MCP-1 contributes to breast cancer. We tested the hypothesis that adipose-produced MCP-1 contributes to mammary tumorigenesis in this study. In a breast cancer model of mouse mammary tumor virus-polyomavirus middle T-antigen (MMTV-PyMT), mice with or without adipose MCP-1 knockout [designated as Mcp-1-/- or wild-type (WT)] were fed the standard AIN93G diet (16% of energy from soybean oil) or a high-fat diet (HFD, 45% of energy from soybean oil). Adipose MCP-1 knockout reduced Mcp-1 expression in adipose tissue and concentrations of MCP-1 in plasma. Mcp-1-/- mice fed the HFD had less body fat than their WT counterparts. Adipose MCP-1 knockout attenuated HFD-enhanced mammary tumorigenesis, evidenced by lower mammary tumor volume. Furthermore, Mcp-1-/- mice, regardless of diet, had a longer tumor latency and less tumor weight than WT mice. When fed the HFD, Mcp-1-/- mice, compared to WT mice, exhibited lower concentrations of insulin, leptin, resistin, vascular endothelial growth factor and hepatic growth factor in plasma. In summary, adipose MCP-1 deficiency attenuated HFD-enhanced MMTV-PyMT mammary tumorigenesis. This attenuation can be attributed to less body adiposity, improvement in insulin sensitivity and down-regulation in protumorigenic inflammation cytokines and angiogenic factors in Mcp-1-/- mice. It concludes that adipose-produced MCP-1 contributes to mammary tumorigenesis in the MMTV-PyMT mouse model.


Subject(s)
Adipose Tissue/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Mammary Neoplasms, Animal/metabolism , Adiposity , Animals , Antigens, Polyomavirus Transforming/metabolism , Chemokine CCL2/blood , Crosses, Genetic , Cytokines/metabolism , Diet, High-Fat , Female , Homozygote , Inflammation , Lung Neoplasms/secondary , Male , Mammary Tumor Virus, Mouse , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic
15.
Int J Mol Sci ; 20(18)2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533313

ABSTRACT

Tumor angiogenesis research and antiangiogenic drug development make use of cultured endothelial cells (ECs) including the human microvascular ECs among others. However, it has been reported that tumor ECs (TECs) are different from normal ECs (NECs). To functionally validate antiangiogenic drugs, cultured TECs are indispensable tools, but are not commercially available. Primary human TECs are available only in small quantities from surgical specimens and have a short life span in vitro due to their cellular senescence. We established immortalized human TECs (h-imTECs) and their normal counterparts (h-imNECs) by infection with lentivirus producing simian virus 40 large T antigen and human telomerase reverse transcriptase to overcome the replication barriers. These ECs exhibited an extended life span and retained their characteristic endothelial morphology, expression of endothelial marker, and ability of tube formation. Furthermore, h-imTECs showed their specific characteristics as TECs, such as increased proliferation and upregulation of TEC markers. Treatment with bevacizumab, an antiangiogenic drug, dramatically decreased h-imTEC survival, whereas the same treatment failed to alter immortalized NEC survival. Hence, these h-imTECs could be a valuable tool for drug screening to develop novel therapeutic agents specific to TECs or functional biological assays in tumor angiogenesis research.


Subject(s)
Cell Transformation, Neoplastic , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kidney Neoplasms/pathology , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Biomarkers , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Ectopic Gene Expression , Humans , Karyotyping , Telomerase/genetics , Telomerase/metabolism
16.
Aging (Albany NY) ; 11(10): 3012-3022, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113906

ABSTRACT

Aging is associated with a genome-wide change of DNA methylation (DNAm). "DNAm age" is defined as the predicted chronological age by the age estimator based on DNAm. The estimator is called the epigenetic clock. The molecular mechanism underlining the epigenetic clock is still unknown. Here, we evaluated the effects of hypoxia and two immortalization factors, hTERT and SV40-LargeT (LT), on the DNAm age of human fibroblasts in vitro. We detected the cell division-associated progression of DNAm age after >10 population doublings. Moreover, the progression of DNAm age was slower under hypoxia (1% oxygen) compared to normoxia (21% oxygen), suggesting that oxygen levels determine the speed of the epigenetic aging. We show that the speed of cell division-associated DNAm age progression depends on the chronological age of the cell donor. hTERT expression did not arrest cell division-associated progression of DNAm age in most cells. SV40LT expression produced inconsistent effects, including rejuvenation of DNAm age. Our results show that a) oxygen and the targets of SV40LT (e.g. p53) modulate epigenetic aging rates and b) the chronological age of donor cells determines the speed of mitosis-associated DNAm age progression in daughter cells.


Subject(s)
Aging/physiology , Biological Clocks , DNA Methylation , Fibroblasts/physiology , Hypoxia/metabolism , Adult , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Epigenesis, Genetic , Humans , Infant, Newborn , Primary Cell Culture , Telomerase/genetics , Telomerase/metabolism
17.
PLoS Pathog ; 15(1): e1007543, 2019 01.
Article in English | MEDLINE | ID: mdl-30689667

ABSTRACT

Merkel cell polyomavirus (MCPyV) accounts for 80% of all Merkel cell carcinoma (MCC) cases through expression of two viral oncoproteins: the truncated large T antigen (LT-t) and small T antigen (ST). MCPyV ST is thought to be the main driver of cellular transformation and has also been shown to increase LT protein levels through the activity of its Large-T Stabilization Domain (LSD). The ST LSD was reported to bind and sequester several ubiquitin ligases, including Fbw7 and ß-TrCP, and thereby stabilize LT-t and several other Fbw7 targets including c-Myc and cyclin E. Therefore, the ST LSD is thought to contribute to transformation by promoting the accumulation of these oncoproteins. Targets of Fbw7 and ß-TrCP contain well-defined, conserved, phospho-degrons. However, as neither MCPyV LT, LT-t nor ST contain the canonical Fbw7 phospho-degron, we sought to further investigate the proposed model of ST stabilization of LT-t and transformation. In this study, we provide several lines of evidence that fail to support a specific interaction between MCPyV T antigens and Fbw7 or ß-TrCP by co-immunoprecipitation or functional consequence. Although MCPyV ST does indeed increase LT protein levels through its Large-T Stabilization domain (LSD), this is accomplished independently of Fbw7. Therefore, our study indicates a need for further investigation into the role and mechanism(s) of MCPyV T antigens in viral replication, latency, transformation, and tumorigenesis.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , F-Box-WD Repeat-Containing Protein 7/metabolism , Merkel cell polyomavirus/metabolism , Antigens, Neoplasm/metabolism , Antigens, Viral, Tumor/metabolism , Carcinoma, Merkel Cell/metabolism , HEK293 Cells , Humans , Ligases/metabolism , Merkel Cells , Merkel cell polyomavirus/immunology , Merkel cell polyomavirus/pathogenicity , Oncogene Proteins/metabolism , Polyomavirus Infections/metabolism , Protein Domains , Tumor Virus Infections/virology , Ubiquitin/metabolism , Virus Replication , beta-Transducin Repeat-Containing Proteins/metabolism
18.
Am J Physiol Renal Physiol ; 316(1): F63-F75, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30303712

ABSTRACT

Glioma-associated oncogene homolog-1 (Gli1)-positive resident mesenchymal stem cell-like cells are the predominant source of kidney myofibroblasts in fibrosis, but investigating Gli1-positive myofibroblast progenitor activation is hampered by the difficulty of isolating and propagating primary cultures of these cells. Using a genetic strategy with positive and negative selection, we isolated Kidney-Gli1 (KGli1) cells that maintain expression of appropriate mesenchymal stem cell-like cell markers, respond to hedgehog pathway activation, and display robust myofibroblast differentiation upon treatment with transforming growth factor-ß (TGF-ß). Coculture of KGli1 cells with endothelium stabilizes capillary formation. Single-cell RNA sequencing (scRNA-seq) analysis during differentiation identified autocrine ligand-receptor pair upregulation and a strong focal adhesion pathway signal. This led us to test the serum response factor inhibitor CCG-203971 that potently inhibited TGF-ß-induced pericyte-to-myofibroblast transition. scRNA-seq also identified the unexpected upregulation of nerve growth factor (NGF), which we confirmed in two mouse kidney fibrosis models. The Ngf receptor Ntrk1 is expressed in tubular epithelium in vivo, suggesting a novel interstitial-to-tubule paracrine signaling axis. Thus, KGli1 cells accurately model myofibroblast activation in vitro, and the development of this cell line provides a new tool to study resident mesenchymal stem cell-like progenitors in health and disease.


Subject(s)
Cell Differentiation , Cell Lineage , Kidney/metabolism , Mesenchymal Stem Cells/metabolism , Myofibroblasts/metabolism , Zinc Finger Protein GLI1/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Cell Line, Transformed , Cell Separation , Coculture Techniques , Epithelial-Mesenchymal Transition , Fibrosis , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kidney/pathology , Mesenchymal Stem Cells/pathology , Mice, Transgenic , Myofibroblasts/pathology , Neovascularization, Physiologic , Paracrine Communication , Phenotype , Signal Transduction , Zinc Finger Protein GLI1/genetics
19.
Breast Cancer Res ; 20(1): 131, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367629

ABSTRACT

BACKGROUND: Amphiregulin (AREG), a ligand of the epidermal growth factor receptor, is not only essential for proper mammary ductal development, but also associated with breast cancer proliferation and growth. In the absence of AREG, mammary ductal growth is stunted and fails to expand. Furthermore, suppression of AREG expression in estrogen receptor-positive breast tumor cells inhibits in-vitro and in-vivo growth. METHODS: We crossed AREG-null (AREG-/-) mice with the murine luminal B breast cancer model, MMTV-PyMT (PyMT), to generate spontaneous breast tumors that lack AREG (AREG-/- PyMT). We evaluated tumor growth, cytokeratin-8 (K8)-positive luminal cells, cytokeratin-14 (K14)-positive myoepithelial cells, and expression of AREG, Ki67, and PyMT. Primary myoepithelial cells from nontumor-bearing AREG+/+ mice underwent fluorescence-activated cell sorting and were adapted to culture for in-vitro coculture studies with AT-3 cells, a cell line derived from C57Bl/6 PyMT mammary tumors. RESULTS: Intriguingly, PyMT-induced lesions progress more rapidly in AREG-/- mice than in AREG+/+ mice. Quantification of K8+ luminal and K14+ myoepithelial cells in non-PyMT AREG-/- mammary glands showed fewer K14+ cells and a thinner myoepithelial layer. Study of AT-3 cells indicated that coculture with myoepithelial cells or exposure to AREG, epidermal growth factor, or basic fibroblast growth factor can suppress PyMT expression. Late-stage AREG-/- PyMT tumors are significantly less solid in structure, with more areas of papillary and cystic growth. Papillary areas appear to be both less proliferative and less necrotic. In The Cancer Genome Atlas database, luminal-B invasive papillary carcinomas have lower AREG expression than luminal B invasive ductal carcinomas. CONCLUSIONS: Our study has revealed a previously unknown role of AREG in myoepithelial cell development and PyMT expression. AREG expression is essential for proper myoepithelial coverage of mammary ducts. Both AREG and myoepithelial cells can suppress PyMT expression. We find that lower AREG expression is associated with invasive papillary breast cancer in both the MMTV-PyMT model and human breast cancer.


Subject(s)
Amphiregulin/metabolism , Epithelial Cells/pathology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Amphiregulin/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/virology , Female , Humans , Mammary Glands, Animal/cytology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness/pathology , Polyomavirus/genetics , Polyomavirus/immunology
20.
PLoS One ; 13(10): e0205297, 2018.
Article in English | MEDLINE | ID: mdl-30296284

ABSTRACT

BACKGROUND: Human ovarian surface epithelial (HOSE) cells are a critical cell source for ovarian cancer research; however, they are difficult to obtain and maintain under standard laboratory conditions in large quantities. The aim of this study was to generate immortalized HOSE (IHOSE) cells with maintained properties to the original cell source, thereby guaranteeing a sufficiently large cell quantity for ovarian cancer research. METHODS: HOSE cells isolated from four non-cancer patients and five IHOSE cell lines were established by induction of HPV-E6/E7 expression or SV40 large T antigen using a lenti-viral system. Each of IHOSE cells was confirmed to be distinct by STR profiling. RNA-sequencing was used to compare gene expression profiles in HOSE, IHOSE and ovarian cancer cells. RESULTS: RNA-sequencing results revealed a stronger linear correlation in gene expression between IHOSE and HOSE cells (R2 = 0.9288) than between IHOSE or HOSE cells and ovarian cancer cells (R2 = 0.8562 and R2 = 0.7982, respectively). The gene expression pattern of 319 differentially expressed genes revealed minimal differences between HOSE and IHOSE cells, while a strong difference between ovarian cancer cells and HOSE or IHOSE cells was observed. Furthermore, the five IHOSE cell lines displayed morphological characteristics typical of epithelial cells but showed a lower level of EpCAM, CD133 and E-cadherin, as cancer stem marker, than ovarian cancer cells. Moreover, unlike cancer cells, IHOSE cells could not form colonies in the anchorage-independent soft agar growth assay. CONCLUSION: These findings demonstrate that five newly established IHOSE cell lines have characteristics of progenitor HOSE cells while exhibiting continuous growth, and thus, should be highly useful as control cells for ovarian cancer research.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Biomarkers, Tumor/genetics , Epithelial Cells/metabolism , Founder Effect , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics , AC133 Antigen/genetics , AC133 Antigen/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Polyomavirus Transforming/metabolism , Biomarkers, Tumor/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Epithelial Cells/cytology , Female , Gene Expression , Humans , Oncogene Proteins, Viral/metabolism , Ovary/cytology , Ovary/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL
...