Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 160
Filter
1.
Viruses ; 16(7)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39066286

ABSTRACT

The BK polyomavirus (BKPyV) is a small DNA non-enveloped virus whose infection is asymptomatic in most of the world's adult population. However, in cases of immunosuppression, the reactivation of the virus can cause various complications, and in particular, nephropathies in kidney transplant recipients or hemorrhagic cystitis in bone marrow transplant recipients. Recently, it was demonstrated that BKPyV virions can use extracellular vesicles to collectively traffic in and out of cells, thus exiting producing cells without cell lysis and entering target cells by diversified entry routes. By a comparison to other naked viruses, we investigated the possibility that BKPyV virions recruit the Endosomal-Sorting Complexes Required for Transport (ESCRT) machinery through late domains in order to hijack extracellular vesicles. We identified a single potential late domain in the BKPyV structural proteins, a YPX3L motif in the VP1 protein, and used pseudovirions to study the effect of point mutations found in a BKPyV clinical isolate or known to ablate the interaction of such a domain with the ESCRT machinery. Our results suggest that this domain is not involved in BKPyV association with extracellular vesicles but is crucial for capsomere interaction and thus viral particle assembly.


Subject(s)
Amino Acid Motifs , BK Virus , Capsid Proteins , Extracellular Vesicles , Virion , Virus Assembly , BK Virus/genetics , BK Virus/physiology , BK Virus/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/virology , Humans , Capsid Proteins/metabolism , Capsid Proteins/genetics , Capsid Proteins/chemistry , Virion/metabolism , Virion/genetics , Polyomavirus Infections/virology , Polyomavirus Infections/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , HEK293 Cells
2.
PLoS Pathog ; 20(7): e1012170, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39074144

ABSTRACT

While Merkel cell polyomavirus (MCPyV or MCV) is an abundant virus frequently shed from healthy skin, it is one of the most lethal tumor viruses in immunocompromised individuals, highlighting the crucial role of host immunity in controlling MCPyV oncogenic potential. Despite its prevalence, very little is known about how MCPyV interfaces with the host immune response to maintain asymptomatic persistent infection and how inadequate control of MCPyV infection triggers MCC tumorigenesis. In this study, we discovered that the MCPyV protein, known as the Alternative Large Tumor Open Reading Frame (ALTO), also referred to as middle T, effectively primes and activates the STING signaling pathway. It recruits Src kinase into the complex of STING downstream kinase TBK1 to trigger its autophosphorylation, which ultimately activates the subsequent antiviral immune response. Combining single-cell analysis with both loss- and gain-of-function studies of MCPyV infection, we demonstrated that the activity of ALTO leads to a decrease in MCPyV replication. Thus, we have identified ALTO as a crucial viral factor that modulates the STING-TBK1 pathway, creating a negative feedback loop that limits viral infection and maintains a delicate balance with the host immune system. Our study reveals a novel mechanism by which a tumorigenic virus-encoded protein can link Src function in cell proliferation to the activation of innate immune signaling, thereby controlling viral spread, and sustaining persistent infection. Our previous findings suggest that STING also functions as a tumor suppressor in MCPyV-driven oncogenesis. This research provides a foundation for investigating how disruptions in the finely tuned virus-host balance, maintained by STING, could alter the fate of MCPyV infection, potentially encouraging malignancy.


Subject(s)
Merkel cell polyomavirus , Polyomavirus Infections , Protein Serine-Threonine Kinases , Tumor Virus Infections , Protein Serine-Threonine Kinases/metabolism , Polyomavirus Infections/metabolism , Polyomavirus Infections/immunology , Polyomavirus Infections/virology , Humans , Tumor Virus Infections/metabolism , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Carcinoma, Merkel Cell/virology , Carcinoma, Merkel Cell/metabolism , Membrane Proteins/metabolism , Signal Transduction , Viral Proteins/metabolism , Virus Replication , Skin Neoplasms/virology , Skin Neoplasms/metabolism , Skin Neoplasms/immunology , Animals
3.
Appl Immunohistochem Mol Morphol ; 31(6): 371-378, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37126387

ABSTRACT

BACKGROUND: Merkel cell carcinoma (MCC) is a rare, aggressive, primary neuroendocrine carcinoma of the skin whose main risk factors are immunosuppression, UV radiation exposure, and Merkel cell polyomavirus. Programmed death-1/programmed death ligand-1 (PD-L1)-based immunotherapy is currently the first choice for treating patients with metastatic MCC. METHODS: MCC biopsies (17) were evaluated for their nucleus and cytoplasm characteristics and growth patterns, as well as for intratumor lymphocytes, mitotic number, and lymphovascular invasion. Paraffin-embedded tissue samples of the biopsies were stained with MCPyV large T-antigen (LTag), RB1, p53, and PD-L1. RESULTS: We observed MCPyV LTag expression in 9 out of the 17 tumors, and all 9 cases were positive for RB1 ( P <0.000). p53 staining was not significantly correlated with MCPyV LTag. We observed no relationship between p53 expression and any other parameters, and PD-L1 expression was low in the MCC samples. We evaluated PD-L1 using both the combined positive score and tumor proportion score (TPS), and found that TPS was correlated with MCPyV LTag expression ( P =0.016). Tumors with tumor-infiltrating lymphocytes showed a better prognosis than those without these lymphocytes ( P =0.006). DISCUSSION: Our data demonstrated that RB1 was effective for immunohistochemically investigating the MCPyV status of tumors. TPS was superior to the combined positive score in evaluating PD-L1 in MCC. Tumor-infiltrating lymphocytes were the only parameters that were associated with survival. Further studies with larger series are required to confirm these results.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Humans , Carcinoma, Merkel Cell/pathology , B7-H1 Antigen/metabolism , Merkel cell polyomavirus/metabolism , Tumor Suppressor Protein p53 , Skin Neoplasms/pathology , Polyomavirus Infections/complications , Polyomavirus Infections/metabolism , Ubiquitin-Protein Ligases , Retinoblastoma Binding Proteins/metabolism
4.
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
5.
Infection ; 51(4): 967-980, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36512270

ABSTRACT

PURPOSE: BK Polyomavirus (BKPyV) infection manifests as renal inflammation and can cause kidney damage. Tumor necrosis factor-α (TNF-α) is increased in renal inflammation and injury. The aim of this study was to investigate the effect of TNF-α blockade on BKPyV infection. METHODS: Urine specimens from 22 patients with BKPyV-associated nephropathy (BKPyVN) and 35 non-BKPyVN kidney transplant recipients were analyzed. RESULTS: We demonstrated increased urinary levels of TNF-α and its receptors, TNFR1 and TNFR2, in BKPyVN patients. Treating BKPyV-infected human proximal tubular cells (HRPTECs) with TNF-α stimulated the expression of large T antigen and viral capsid protein-1 mRNA and proteins and BKPyV promoter activity. Knockdown of TNFR1 or TNFR2 expression caused a reduction in TNF-α-stimulated viral replication. NF-κB activation induced by overexpression of constitutively active IKK2 significantly increased viral replication and the activity of the BKPyV promoter containing an NF-κB binding site. The addition of a NF-κB inhibitor on BKPyV-infected cells suppressed viral replication. Blockade of TNF-α functionality by etanercept reduced BKPyV-stimulated expression of TNF-α, interleukin-1ß (IL-1ß), IL-6 and IL-8 and suppressed TNF-α-stimulated viral replication. In cultured HRPTECs and THP-1 cells, BKPyV infection led to increased expression of TNF-α, interleukin-1 ß (IL-1ß), IL-6 and TNFR1 and TNFR2 but the stimulated magnitude was far less than that induced by poly(I:C). This may suggest that BKPyV-mediated autocrine effect is not a major source of TNFα. CONCLUSION: TNF-α stimulates BKPyV replication and inhibition of its signal cascade or functionality attenuates its stimulatory effect. Our study provides a therapeutic anti-BKPyV target.


Subject(s)
BK Virus , Polyomavirus Infections , Humans , BK Virus/genetics , Tumor Necrosis Factor-alpha , Receptors, Tumor Necrosis Factor, Type I , Receptors, Tumor Necrosis Factor, Type II/genetics , NF-kappa B , Interleukin-6 , Polyomavirus Infections/metabolism , Polyomavirus Infections/pathology , Inflammation
6.
PLoS Pathog ; 18(12): e1011039, 2022 12.
Article in English | MEDLINE | ID: mdl-36574443

ABSTRACT

Merkel cell polyomavirus (MCV) is a small DNA tumor virus that persists in human skin and causes Merkel cell carcinoma (MCC) in immunocompromised individuals. The multi-functional protein MCV small T (sT) activates viral DNA replication by stabilizing large T (LT) and promotes cell transformation through the LT stabilization domain (LTSD). Using MCVΔsT, a mutant MCV clone that ablates sT, we investigated the role of sT in MCV genome maintenance. sT was dispensable for initiation of viral DNA replication, but essential for maintenance of the MCV genome and activation of viral early and late gene expression for progression of the viral lifecycle. Furthermore, in phenotype rescue studies, exogenous sT activated viral DNA replication and mRNA expression in MCVΔsT through the LTSD. While exogenous LT expression, which mimics LT stabilization, increased viral DNA replication, it did not activate viral mRNA expression. After cataloging transcriptional regulator proteins by proximity-based MCV sT-host protein interaction analysis, we validated LTSD-dependent sT interaction with four transcriptional regulators: Cux1, c-Jun, BRD9, and CBP. Functional studies revealed Cux1 and c-Jun as negative regulators, and CBP and BRD9 as positive regulators of MCV transcription. CBP inhibitor A-485 suppressed sT-induced viral gene activation in replicating MCVΔsT and inhibited early gene expression in MCV-integrated MCC cells. These results suggest that sT promotes viral lifecycle progression by activating mRNA expression and capsid protein production through interaction with the transcriptional regulators. This activity is essential for MCV genome maintenance, suggesting a critical role of sT in MCV persistence and MCC carcinogenesis.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Tumor Virus Infections , Humans , Merkel cell polyomavirus/metabolism , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/metabolism , Viral Transcription , DNA Replication , Virus Replication , DNA, Viral/genetics , DNA, Viral/metabolism , Transcription Factors/metabolism , Skin Neoplasms/pathology , Genome, Viral , RNA, Messenger/metabolism , Polyomavirus Infections/metabolism
7.
Virchows Arch ; 480(6): 1239-1254, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35412101

ABSTRACT

MCC (Merkel cell carcinoma) is an aggressive neuroendocrine cutaneous neoplasm. Integration of the Merkel cell polyomavirus (MCPyV) is observed in about 80% of the cases, while the remaining 20% are related to UV exposure. Both MCPyV-positive and -negative MCCs-albeit by different mechanisms-are associated with RB1 inactivation leading to overexpression of SOX2, a major contributor to MCC biology. Moreover, although controversial, loss of RB1 expression seems to be restricted to MCPyV-negative cases.The aim of the present study was to assess the performances of RB1 loss and SOX2 expression detected by immunohistochemistry to determine MCPyV status and to diagnose MCC, respectively.Overall, 196 MCC tumors, 233 non-neuroendocrine skin neoplasms and 70 extra-cutaneous neuroendocrine carcinomas (NEC) were included. SOX2 and RB1 expressions were assessed by immunohistochemistry in a tissue micro-array. Diagnostic performances were determined using the likelihood ratio (LHR).RB1 expression loss was evidenced in 27% of the MCC cases, 12% of non-neuroendocrine skin tumors and 63% of extra-cutaneous NEC. Importantly, among MCC cases, RB1 loss was detected in all MCPyV(-) MCCs, while MCPyV( +) cases were consistently RB1-positive (p < 0.001). SOX2 diffuse expression was observed in 92% of the MCC cases and almost never observed in non-neuroendocrine skin epithelial neoplasms (2%, p < 0.0001, LHR + = 59). Furthermore, SOX2 diffuse staining was more frequently observed in MCCs than in extra-cutaneous NECs (30%, p < 0.001, LHR + = 3.1).These results confirm RB1 as a robust predictor of MCC viral status and further suggest SOX2 to be a relevant diagnostic marker of MCC.


Subject(s)
Carcinoma, Merkel Cell , Merkel cell polyomavirus , Polyomavirus Infections , Skin Neoplasms , Tumor Virus Infections , Carcinoma, Merkel Cell/diagnosis , Carcinoma, Merkel Cell/pathology , Humans , Merkel cell polyomavirus/metabolism , Polyomavirus Infections/complications , Polyomavirus Infections/metabolism , Retinoblastoma Binding Proteins , SOXB1 Transcription Factors/metabolism , Skin Neoplasms/pathology , Tumor Virus Infections/complications , Ubiquitin-Protein Ligases
8.
J Cutan Pathol ; 49(1): 49-54, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34313332

ABSTRACT

BACKGROUND: Merkel cell polyomavirus (MCPyV) has been studied in several malignant and nonmalignant tissues. However, only in Merkel cell carcinoma (MCC) has the connection to tumorigenesis been established. Previously, eccrine porocarcinoma samples were shown to express MCPyV in the majority of samples. We aimed to examine MCPyV in porocarcinoma and poroma samples using MCC as the reference material. METHODS: We analyzed 17 porocarcinoma and 50 poroma samples for the presence of MCPyV using LT antigen immunostaining and DNA detection methods. In addition, 180 MCC samples served as controls. RESULTS: MCPyV LT antigen immunostaining was detected in 10% of poroma and 18% of porocarcinoma samples; on the other hand, it was present in 65% of MCC samples. MCPyV DNA was detected in only 10% of poroma and porocarcinoma samples compared with 96% of MCC samples. The viral DNA copy number in all MCPyV DNA-positive MCCs was at least 25 times higher than that in porocarcinoma or poroma samples with the highest MCPyV DNA-to-PTPRG ratio. CONCLUSIONS: The low number of viral DNA copies in poroma and porocarcinoma samples, together with the negative LT expression of MCPyV DNA-positive tumors, indicates that MCPyV is simply a passenger virus rather than an oncogenic driver of porocarcinoma.


Subject(s)
Carcinoma, Merkel Cell , Eccrine Porocarcinoma , Merkel cell polyomavirus/metabolism , Polyomavirus Infections , Sweat Gland Neoplasms , Tumor Virus Infections , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Carcinoma, Merkel Cell/virology , Child , Eccrine Porocarcinoma/metabolism , Eccrine Porocarcinoma/pathology , Eccrine Porocarcinoma/virology , Female , Humans , Male , Middle Aged , Polyomavirus Infections/metabolism , Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Sweat Gland Neoplasms/metabolism , Sweat Gland Neoplasms/pathology , Sweat Gland Neoplasms/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
9.
PLoS Pathog ; 17(2): e1009042, 2021 02.
Article in English | MEDLINE | ID: mdl-33592065

ABSTRACT

The BK polyomavirus (BKPyV) is a ubiquitous human virus that persists in the renourinary epithelium. Immunosuppression can lead to BKPyV reactivation in the first year post-transplantation in kidney transplant recipients (KTRs) and hematopoietic stem cell transplant recipients. In KTRs, persistent DNAemia has been correlated to the occurrence of polyomavirus-associated nephropathy (PVAN) that can lead to graft loss if not properly controlled. Based on recent observations that conventional dendritic cells (cDCs) specifically infiltrate PVAN lesions, we hypothesized that those cells could play a role in BKPyV infection. We first demonstrated that monocyte-derived dendritic cells (MDDCs), an in vitro model for mDCs, captured BKPyV particles through an unconventional GRAF-1 endocytic pathway. Neither BKPyV particles nor BKPyV-infected cells were shown to activate MDDCs. Endocytosed virions were efficiently transmitted to permissive cells and protected from the antibody-mediated neutralization. Finally, we demonstrated that freshly isolated CD1c+ mDCs from the blood and kidney parenchyma behaved similarly to MDDCs thus extending our results to cells of clinical relevance. This study sheds light on a potential unprecedented CD1c+ mDC involvement in the BKPyV infection as a promoter of viral spreading.


Subject(s)
Antigens, CD1/metabolism , BK Virus/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Glycoproteins/metabolism , Kidney/immunology , Polyomavirus Infections/immunology , Tumor Virus Infections/immunology , Antibodies, Neutralizing/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Kidney/metabolism , Kidney/virology , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Polyomavirus Infections/metabolism , Polyomavirus Infections/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Virus Replication
10.
Pediatr Transplant ; 25(2): e13894, 2021 03.
Article in English | MEDLINE | ID: mdl-33136312

ABSTRACT

The aims were to investigate the incidence of BKV infection and the presence of HC in pediatric patients undergoing HSCT. Twenty-four children patients (M/F: 17/7) undergoing HSCT in a single center over a period of 1 year were included in the study. The presence of BKV DNA was determined by quantitative real-time PCR in plasma and urine samples at the following times: before transplantation, twice a week until engraftment time, and weekly for + 100 days. The mean age of the patients was 7.79 ± 5.03 years, the mean follow-up time was 95.6 ± 25.9 days, and the average number of samples per patient was 15.8 ± 3.2. BKV DNA was detected in at least one urine sample in 91.6% (n: 22) and at least one plasma sample in 75% (n:18) of the patients. The median time to the first BKV DNA positivity in urine and plasma samples was 11 (range: 1-80) and 32 days (range: 2-79), respectively. The median value of BKV DNA copies in urine and plasma were 1.7 × 106 (range: 2.8 × 101 -1.2 × 1014 ) and 1.9 × 103 copies/mL (range: 3-2.1 × 106 ), respectively. Thirteen patients (54.2%) had hematuria with BKV viruria; 8 (33.3%) patients had viremia. The median value of the BKV DNA copies in urine and plasma was 4.4 × 107 (range: 65-1 × 1011 ) and 2.9 × 103 (range: 7-7.8 × 104 ) copies/mL in these patients. Two (15.4%) of the 13 patients with BKV viruria and hematuria were diagnosed with BKV-related HC. BKV DNA viral load monitoring of urine and plasma in pediatric HSCT patients with a high risk for viral infections is valuable for understanding the development of BKV-related HC.


Subject(s)
BK Virus/isolation & purification , Cystitis/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Immunocompromised Host , Immunosuppressive Agents/adverse effects , Polyomavirus Infections/immunology , Adolescent , Child , Child, Preschool , Cystitis/diagnosis , Cystitis/epidemiology , Cystitis/virology , Female , Follow-Up Studies , Humans , Incidence , Male , Polyomavirus Infections/diagnosis , Polyomavirus Infections/epidemiology , Polyomavirus Infections/metabolism , Viral Load , Young Adult
11.
Viruses ; 12(12)2020 12 09.
Article in English | MEDLINE | ID: mdl-33317205

ABSTRACT

BK virus (BKV) is a polyomavirus with high seroprevalence in the general population with an unremarkable clinical presentation in healthy people, but a potential for causing serious complications in immunosuppressed transplanted patients. Reactivation or primary infection in kidney allograft recipients may lead to allograft dysfunction and subsequent loss. Currently, there is no widely accepted specific treatment for BKV infection and reduction of immunosuppressive therapy is the mainstay therapy. Given this and the sequential appearance of viruria-viremia-nephropathy, screening and early detection are of utmost importance. There are numerous risk factors associated with BKV infection including genetic factors, among them human leukocyte antigens (HLA) and killer cell immunoglobulin-like receptors (KIR) alleles have been shown to be the strongest so far. Identification of patients at risk for BKV infection would be useful in prevention or early action to reduce morbidity and progression to frank nephropathy. Assessment of risk involving HLA ligands and KIR genotyping of recipients in the pre-transplant or early post-transplant period might be useful in clinical practice. This review summarizes current knowledge of the association between HLA, KIR and BKV infection and potential future directions of research, which might lead to optimal utilization of these genetic markers.


Subject(s)
BK Virus/physiology , HLA Antigens/immunology , Kidney Transplantation/adverse effects , Polyomavirus Infections/etiology , Polyomavirus Infections/metabolism , Receptors, KIR/metabolism , Alleles , Biomarkers , Disease Susceptibility , HLA Antigens/genetics , Humans , Immunocompromised Host , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Prognosis , Research
12.
JCI Insight ; 5(22)2020 11 19.
Article in English | MEDLINE | ID: mdl-33055416

ABSTRACT

The challenge of discovering a completely new human tumor virus of unknown phylogeny or sequence depends on detecting viral molecules and differentiating them from host molecules in the virus-associated neoplasm. We developed differential peptide subtraction (DPS) using differential mass spectrometry (dMS) followed by targeted analysis to facilitate this discovery. We validated this approach by analyzing Merkel cell carcinoma (MCC), an aggressive human neoplasm, in which ~80% of cases are caused by the human Merkel cell polyomavirus (MCV). Approximately 20% of MCC have a high mutational burden and are negative for MCV, but are microscopically indistinguishable from virus positive cases. Using 23 (12 MCV+, 11 MCV-) formalin-fixed MCC, DPS identified both viral and human biomarkers (MCV large T antigen, CDKN2AIP, SERPINB5, and TRIM29) that discriminate MCV+ and MCV- MCC. Statistical analysis of 498,131 dMS features not matching the human proteome by DPS revealed 562 (0.11%) to be upregulated in virus-infected samples. Remarkably, 4 (20%) of the top 20 candidate MS spectra originated from MCV T oncoprotein peptides and confirmed by reverse translation degenerate oligonucleotide sequencing. DPS is a robust proteomic approach to identify potentially novel viral sequences in infectious tumors when nucleic acid-based methods are not feasible.


Subject(s)
Antigens, Viral, Tumor/metabolism , Biomarkers/metabolism , Carcinoma, Merkel Cell/diagnosis , Polyomavirus Infections/complications , Proteome/metabolism , Skin Neoplasms/diagnosis , Tumor Virus Infections/complications , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Carcinoma, Merkel Cell/virology , Formaldehyde/chemistry , Humans , Merkel cell polyomavirus/physiology , Polyomavirus Infections/metabolism , Polyomavirus Infections/virology , Proteome/analysis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology
13.
Viruses ; 12(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-33028008

ABSTRACT

Polyomaviruses are a family of small, non-enveloped DNA viruses that can cause severe disease in immunosuppressed individuals. Studies with SV40, a well-studied model polyomavirus, have revealed the role of host proteins in polyomavirus entry and trafficking to the nucleus, in viral transcription and DNA replication, and in cell transformation. In contrast, little is known about host factors or cellular signaling pathways involved in the late steps of productive infection leading to release of progeny polyomaviruses. We previously showed that cytoplasmic vacuolization, a characteristic late cytopathic effect of SV40 infection, depends on the specific interaction between the major viral capsid protein VP1 and its cell surface ganglioside receptor GM1. Here, we show that, late during infection, SV40 activates a signaling cascade in permissive monkey CV-1 cells involving Ras, Rac1, MKK4, and JNK to stimulate SV40-specific cytoplasmic vacuolization and subsequent cell lysis and virus release. Inhibition of individual components of this signaling pathway inhibits vacuolization, lysis, and virus release, even though high-level intracellular virus replication occurs. Identification of this pathway for SV40-induced vacuolization and virus release provides new insights into the late steps of non-enveloped virus infection.


Subject(s)
Cell Death/physiology , MAP Kinase Signaling System/physiology , Simian virus 40/metabolism , Virus Release/physiology , Capsid Proteins , Cell Line , Humans , MAP Kinase Kinase 4/metabolism , Polyomavirus Infections/metabolism , Simian virus 40/genetics , Tumor Virus Infections/metabolism , rac1 GTP-Binding Protein/metabolism
14.
Virol J ; 17(1): 139, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32928222

ABSTRACT

BACKGROUND: Recent studies have confirmed the integration of the BK polyomavirus (BKPyV) gene into the cellular genome of urothelial carcinomas in transplant recipients, further confirming the correlation between BKPyV and urothelial carcinomas after transplantation. However, the role BKPyV infections play in the biological function of bladder cancer remains unclear. METHODS: We developed a BKPyV-infected bladder cancer cell model and a mice tumor model to discuss the role of BKPyV infections. RESULTS: Our research proves that BKPyV infections promote the proliferation, invasion and migration of bladder cancer cells, while the activation of ß-catenin signaling pathway is one of its mediation mechanisms. CONCLUSIONS: We first described BKPyV infection promotes the proliferation, invasion and migration of bladder cancer. We verified the role of ß-catenin signaling pathway and Epithelial-Mesenchymal Transition effect in BKPyV-infected bladder cancer. These results provide meaningful information towards the diagnosis and treatment of clinical bladder cancer.


Subject(s)
BK Virus/pathogenicity , Polyomavirus Infections/pathology , Tumor Virus Infections/pathology , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Disease Models, Animal , Epithelial-Mesenchymal Transition , Humans , Male , Mice , Mice, Inbred BALB C , Polyomavirus Infections/metabolism , Polyomavirus Infections/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/virology , Wnt Signaling Pathway , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
15.
Viruses ; 12(9)2020 08 31.
Article in English | MEDLINE | ID: mdl-32878339

ABSTRACT

Merkel cell carcinoma (MCC) is an uncommon, lethal cancer of the skin caused by either Merkel cell polyomavirus (MCPyV) or UV-linked mutations. MCPyV is found integrated into MCC tumor genomes, accompanied by truncation mutations that render the MCPyV large T antigen replication incompetent. We used the open access HPV Detector/Cancer-virus Detector tool to determine MCPyV integration sites in whole-exome sequencing data from five MCC cases, thereby adding to the limited published MCPyV integration site junction data. We also systematically reviewed published data on integration for MCPyV in the human genome, presenting a collation of 123 MCC cases and their linked chromosomal sites. We confirmed that there were no highly recurrent specific sites of integration. We found that chromosome 5 was most frequently involved in MCPyV integration and that integration sites were significantly enriched for genes with binding sites for oncogenic transcription factors such as LEF1 and ZEB1, suggesting the possibility of increased open chromatin in these gene sets. Additionally, in one case we found, for the first time, integration involving the tumor suppressor gene KMT2D, adding to previous reports of rare MCPyV integration into host tumor suppressor genes in MCC.


Subject(s)
Carcinoma, Merkel Cell/metabolism , DNA-Binding Proteins/metabolism , Merkel cell polyomavirus/physiology , Neoplasm Proteins/metabolism , Polyomavirus Infections/metabolism , Tumor Virus Infections/metabolism , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/virology , Cell Line, Tumor , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , Chromosomes, Human, Pair 5/virology , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Humans , Merkel cell polyomavirus/genetics , Neoplasm Proteins/genetics , Polyomavirus Infections/genetics , Polyomavirus Infections/virology , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , Virus Integration , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
16.
Virology ; 548: 124-131, 2020 09.
Article in English | MEDLINE | ID: mdl-32838933

ABSTRACT

SP1 binding in SV40 chromatin in vitro and in vivo was characterized in order to better understand its role during the initiation of early transcription. We observed that chromatin from disrupted virions, but not minichromosomes, was efficiently bound by HIS-tagged SP1 in vitro, while the opposite was true for the presence of endogenous SP1 introduced in vivo. Using ChIP-Seq to compare the location of SP1 to nucleosomes carrying modified histones, we found that SP1 could occupy its whole binding site in virion chromatin but only the early side of its binding site in most of the minichromosomes carrying modified histones due to the presence of overlapping nucleosomes. The results suggest that during the initiation of an SV40 infection, SP1 binds to an open region in SV40 virion chromatin but quickly triggers chromatin reorganization and its own removal.


Subject(s)
Chromatin/virology , Polyomavirus Infections/metabolism , Polyomavirus Infections/virology , Simian virus 40/metabolism , Sp1 Transcription Factor/metabolism , Virion/metabolism , Chromatin/genetics , Chromatin/metabolism , Host-Pathogen Interactions , Humans , Nucleosomes/genetics , Nucleosomes/metabolism , Polyomavirus Infections/genetics , Protein Binding , Simian virus 40/genetics , Sp1 Transcription Factor/genetics , Virion/genetics
17.
Virology ; 548: 17-24, 2020 09.
Article in English | MEDLINE | ID: mdl-32838939

ABSTRACT

The demyelinating disease progressive multifocal leukoencephalopathy (PML) is caused by the human polyomavirus, JCPyV, under conditions of prolonged immunosuppression. Initial infection is asymptomatic, and the virus establishes lifelong persistence in the host. Following the loss of immune surveillance, the virus can traffic to the central nervous system and infect oligodendrocytes to cause demyelination and PML. The mechanisms involved in glial cell infection are not completely understood. In a screen for N-glycosylated proteins that influence JCPyV pathology, we identified Adipocyte Plasma Membrane Associated Protein (APMAP) as a host cell modulator of JCPyV infection. The removal of APMAP by small interfering siRNA as well as by CRISPR-Cas9 gene editing resulted in a significant decrease in JCPyV infection. Exogenous expression of APMAP in APMAP knockout cell lines rescued susceptibility to infection. These data suggest that virus infection of glial cells is dependent on APMAP.


Subject(s)
JC Virus/physiology , Neuroglia/metabolism , Polyomavirus Infections/metabolism , Cell Line , Host-Pathogen Interactions , Humans , JC Virus/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins , Neuroglia/virology , Oligodendroglia/metabolism , Oligodendroglia/virology , Polyomavirus Infections/genetics , Polyomavirus Infections/virology
18.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32669331

ABSTRACT

Merkel cell polyomavirus (MCV) small T antigen (sT) is the main oncoprotein for the development of Merkel cell carcinoma (MCC). MCC is a rare, clinically aggressive neuroendocrine tumor of the skin with a high propensity for local, regional, and distant spread. The dysregulation of matrix metalloproteinase-9 (MMP-9) has been implicated in multiple essential roles in the development of various malignant tumor cell invasion and metastasis. Previously, MCV sT was shown to induce the migratory and invasive phenotype of MCC cells through the transcriptional activation of the sheddase molecule, ADAM 10 (A disintegrin and metalloprotease domain-containing protein 10). In this study, we show that MCV sT protein stimulates differential expression of epithelial-mesenchymal transition (EMT)-associated genes, including MMP-9 and Snail. This effect is dependent on the presence of the large T stabilization domain (LSD), which is known to be responsible for cell transformation through targeting of promiscuous E3 ligases, including FBW7, a known MMP-9 and Snail regulator. Chemical treatments of MMP-9 markedly inhibited MCV sT-induced cell migration and invasion. These results suggest that MCV sT contributes to the activation of MMP-9 as a result of FBW7 targeting and increases the invasive potential of cells, which can be used for targeted therapeutic intervention.IMPORTANCE Merkel cell carcinoma (MCC) is the most aggressive cutaneous tumor without clearly defined treatment. Although MCC has a high propensity for metastasis, little is known about the underlying mechanisms that drive MCC invasion and metastatic progression. MMP-9 has been shown to play a detrimental role in many metastatic human cancers, including melanoma and other nonmelanoma skin cancers. Our study shows that MCV sT-mediated MMP-9 activation is driven through the LSD, a known E3 ligase-targeting domain, in MCC. MMP-9 may serve as the biochemical culprit to target and develop a novel approach for the treatment of metastatic MCC.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Movement/genetics , Cell Movement/physiology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Merkel cell polyomavirus/metabolism , ADAM10 Protein/genetics , ADAM10 Protein/metabolism , Animals , Antigens, Viral, Tumor , COS Cells , Carcinoma, Merkel Cell/virology , Cell Proliferation , Cell Transformation, Neoplastic , Chlorocebus aethiops , Epithelial-Mesenchymal Transition/genetics , Gene Expression , HEK293 Cells , Humans , Oncogene Proteins , Polyomavirus Infections/metabolism , Skin Neoplasms/virology , Snails , Tumor Virus Infections/virology , Ubiquitin-Protein Ligases/metabolism
19.
J Cell Mol Med ; 24(16): 9097-9100, 2020 08.
Article in English | MEDLINE | ID: mdl-32588533

ABSTRACT

The BK polyomavirus (BKPyV) has pathogenic relevance especially in immunocompromised patients. No causal therapy has been established yet. Therefore, new therapeutic targets need to be identified in experimental studies. A 3D organotypic cell culture model with primary urothelial cells and fibroblasts was used as infection model. The detection of virus replication was performed with quantitative polymerase chain reaction (qPCR), and immunohistochemistry (IHC) was also used for analysis. Interleukin levels were measured by enzyme-linked immunosorbent assay (ELISA). Interestingly, the signal transducer and activator of transcription 3 (STAT3) pathway seems to be activated during infection with BKPyV, for example phosphorylated STAT3 is significantly (P < 0.0001) elevated on day 6 following infection. Therefore, we performed ELISAs for involved interleukins in STAT3 pathway. Interleukin 11 (IL-11) was significantly (P = 0.026) elevated at day 9. Subsequently, 3D cultures were treated with IL-11 neutralizing antibody. At day 9 following infection, the median virus replication rate is 4.4 × 106 copies/ml. The difference to replication rate without treatment was significantly lower at day 6 (P < 0.0001) and at day 9 (P < 0.0001), respectively. STAT3 pathways seem to be involved during BKPyV infection and need further investigation in experimental studies. A very promising target for treatment might be IL-11.


Subject(s)
BK Virus/pathogenicity , Hemorrhage/metabolism , Interleukin-11/metabolism , Polyomavirus Infections/metabolism , BK Virus/genetics , Cell Culture Techniques/methods , Cells, Cultured , Cystitis , Fibroblasts/metabolism , Fibroblasts/virology , Hemorrhage/virology , Humans , Polyomavirus Infections/virology , STAT3 Transcription Factor/metabolism , Urothelium/metabolism , Urothelium/virology , Virus Replication/genetics
20.
J Virol ; 94(11)2020 05 18.
Article in English | MEDLINE | ID: mdl-32161173

ABSTRACT

The nonenveloped polyomavirus simian virus 40 (SV40) must penetrate the host endoplasmic reticulum (ER) membrane to enter the cytosol in order to promote infection. How this is accomplished is not entirely clear. Here, we demonstrate that the cytosolic chaperone Ubiquilin4 (Ubqln4) binds directly to the ER membrane J proteins B12 and B14. Strategically localized at the ER-cytosol interface, Ubqln4 captures SV40 emerging from the ER, thereby facilitating escape of the virus from the ER into the cytosol, which leads to infection. Strikingly, Ubqln4 engages the J proteins in a J-domain-independent manner, in contrast to the previously reported Hsc70-Hsp105-SGTA-Bag2 cytosolic complex that also mediates SV40 ER-to-cytosol transport. Our results also reveal that the H domain and STI1 motif (1-2) of Ubqln4 support J protein binding, essential for SV40 infection. Together, these data further clarify the molecular basis by which a nonenveloped virus escapes a host membrane during infectious entry.IMPORTANCE How a nonenveloped virus escapes from a host membrane to promote infection remains enigmatic. In the case of the nonenveloped polyomavirus SV40, penetration of the ER membrane to reach the cytosol is a decisive virus infection step. In this study, we found a new host factor called Ubqln4 that facilitates escape of SV40 from the ER into the cytosol, thereby providing a path for the virus to enter the nucleus to cause infection.


Subject(s)
Carrier Proteins/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Nuclear Proteins/metabolism , Polyomavirus Infections/metabolism , Simian virus 40/metabolism , Amino Acid Motifs , Biological Transport, Active/genetics , Carrier Proteins/genetics , Cell Line , Cytosol/pathology , Cytosol/virology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/virology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Polyomavirus Infections/genetics , Polyomavirus Infections/pathology , Protein Domains , Simian virus 40/genetics
SELECTION OF CITATIONS
SEARCH DETAIL