Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Infect Dis ; 224(7): 1160-1169, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32060513

RESUMO

BACKGROUND: Human polyomaviruses can reactivate in transplant patients, causing nephropathy, progressive multifocal leukoencephalopathy, Merkel cell carcinoma, pruritic, rash or trichodysplasia spinulosa. Sirolimus and related mechanistic target of rapamycin (mTOR) inhibitors are transplant immunosuppressants. It is unknown if they directly reactivate polyomavirus replication from latency beyond their general effects on immunosuppression. METHODS: In vitro expression and turnover of large T (LT) proteins from BK virus, JC virus (JCV), Merkel cell polyomavirus (MCV), human polyomavirus 7 (HPyV7), and trichodysplasia spinulosa polyomavirus (TSV) after drug treatment were determined by immunoblotting, proximity ligation, replicon DNA replication, and whole virus immunofluorescence assays. RESULTS: mTOR inhibition increased LT protein expression for all 5 pathogenic polyomaviruses tested. This correlated with LT stabilization, decrease in the S-phase kinase-associated protein 2 (Skp2) E3 ligase targeting these LT proteins for degradation, and increase in virus replication for JCV, MCV, TSV, and HPyV7. Treatment with sirolimus, but not the calcineurin inhibitor tacrolimus, at levels routinely achieved in patients, resulted in a dose-dependent increase in viral DNA replication for BKV, MCV, and HPyV7. CONCLUSIONS: mTOR inhibitors, at therapeutic levels, directly activate polyomavirus replication through a Skp2-dependent mechanism, revealing a proteostatic latency mechanism common to polyomaviruses. Modifying existing drug regimens for transplant patients with polyomavirus-associated diseases may reduce symptomatic polyomavirus replication while maintaining allograft-sparing immunosuppression.


Assuntos
Replicação do DNA/efeitos dos fármacos , Inibidores de MTOR/farmacologia , Polyomavirus/efeitos dos fármacos , Proteínas Quinases Associadas a Fase S , Sirolimo/farmacologia , Replicação Viral/efeitos dos fármacos , Vírus BK , DNA Viral , Humanos , Vírus JC , Poliomavírus das Células de Merkel , Polyomavirus/genética , Infecções por Polyomavirus/tratamento farmacológico , Serina-Treonina Quinases TOR , Tacrolimo/farmacologia
2.
J Virol ; 94(19)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32669331

RESUMO

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.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Movimento Celular/genética , Movimento Celular/fisiologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Poliomavírus das Células de Merkel/metabolismo , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Animais , Antígenos Virais de Tumores , Células COS , Carcinoma de Célula de Merkel/virologia , Proliferação de Células , Transformação Celular Neoplásica , Chlorocebus aethiops , Transição Epitelial-Mesenquimal/genética , Expressão Gênica , Células HEK293 , Humanos , Proteínas Oncogênicas , Infecções por Polyomavirus/metabolismo , Neoplasias Cutâneas/virologia , Caramujos , Infecções Tumorais por Vírus/virologia , Ubiquitina-Proteína Ligases/metabolismo
3.
Int J Mol Sci ; 22(13)2021 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-34281220

RESUMO

Merkel cell polyomavirus (MCPyV) large tumor (LT) antigen is a DNA binding protein essential for viral gene transcription and genome replication. MCPyV LT interacts with multiple E3 ligases in a phosphorylation-dependent manner, limiting its own viral replication by enhancing LT protein degradation, which is a unique mechanism for MCPyV latency. Thus, identifying LT ubiquitination sites is an important step toward understanding the biological role of these virus-host interactions that can potentially result in viral oncogenesis. The ubiquitin (Ub) attachment sites in LT were predicted by using Rapid UBIquitination (RUBI), a sequence-based ubiquitination web server. Using an immunoprecipitation approach, the lysine (Lys, K) 585 residue in LT is identified as the ubiquitin conjugation site. Lysine 585 is deleted from tumor-derived truncated LTs (tLTs), resulting in stable expression of tLTs present in cancers. Substitution of lysine 585 to arginine (Arg, R) increased LT protein stability, but impaired MCPyV origin replication, due to a loss of ATP hydrolysis activity. These findings uncover a never-before-identified ubiquitination site of LT and its importance not only in the regulation of protein turnover, but also in MCPyV genome replication.


Assuntos
Antígenos Virais de Tumores/metabolismo , Poliomavírus das Células de Merkel/imunologia , Trifosfato de Adenosina/metabolismo , Células HEK293 , Humanos , Poliomavírus das Células de Merkel/metabolismo , Estabilidade Proteica , Ubiquitinação , Replicação Viral
4.
Proc Natl Acad Sci U S A ; 114(20): E4040-E4047, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461484

RESUMO

Viral latency, in which a virus genome does not replicate independently of the host cell genome and produces no infectious particles, is required for long-term virus persistence. There is no known latency mechanism for chronic small DNA virus infections. Merkel cell polyomavirus (MCV) causes an aggressive skin cancer after prolonged infection and requires an active large T (LT) phosphoprotein helicase to replicate. We show that evolutionarily conserved MCV LT phosphorylation sites are constitutively recognized by cellular Fbw7, ßTrCP, and Skp2 Skp-F-box-cullin (SCF) E3 ubiquitin ligases, which degrade and suppress steady-state LT protein levels. Knockdown of each of these E3 ligases enhances LT stability and promotes MCV genome replication. Mutations at two of these phosphoreceptor sites [serine (S)220 and S239] in the full viral genome increase LT levels and promote MCV virion production and transmission, which can be neutralized with anti-capsid antibody. Virus activation is not mediated by viral gene transactivation, given that these mutations do not increase late gene transcription in the absence of genome replication. Mechanistic target of rapamycin inhibition by either nutrient starvation or use of an active site inhibitor reduces Skp2 levels and stabilizes LT, leading to enhanced MCV replication and transmission. MCV can sense stresses in its intracellular environment, such as nutrient loss, through SCF E3 ligase activities, and responds by initiating active viral transmission. Protein-mediated viral latency through cellular SCF E3 ligase targeting of viral replication proteins is a unique form of latency that may promote chronic viral persistence for some small DNA and RNA viruses.


Assuntos
Antígenos Transformantes de Poliomavirus/metabolismo , Poliomavírus das Células de Merkel/fisiologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Latência Viral , Células HEK293 , Humanos , Replicação Viral
5.
Proc Natl Acad Sci U S A ; 112(19): 5875-82, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25883264

RESUMO

Mitosis is commonly thought to be associated with reduced cap-dependent protein translation. Here we show an alternative control mechanism for maintaining cap-dependent translation during mitosis revealed by a viral oncoprotein, Merkel cell polyomavirus small T (MCV sT). We find MCV sT to be a promiscuous E3 ligase inhibitor targeting the anaphase-promoting complex, which increases cell mitogenesis. MCV sT binds through its Large T stabilization domain region to cell division cycle protein 20 (Cdc20) and, possibly, cdc20 homolog 1 (Cdh1) E3 ligase adapters. This activates cyclin-dependent kinase 1/cyclin B1 (CDK1/CYCB1) to directly hyperphosphorylate eukaryotic initiation factor 4E (eIF4E)-binding protein (4E-BP1) at authentic sites, generating a mitosis-specific, mechanistic target of rapamycin (mTOR) inhibitor-resistant δ phospho-isoform not present in G1-arrested cells. Recombinant 4E-BP1 inhibits capped mRNA reticulocyte translation, which is partially reversed by CDK1/CYCB1 phosphorylation of 4E-BP1. eIF4G binding to the eIF4E-m(7)GTP cap complex is resistant to mTOR inhibition during mitosis but sensitive during interphase. Flow cytometry, with and without sT, reveals an orthogonal pH3(S10+) mitotic cell population having higher inactive p4E-BP1(T37/T46+) saturation levels than pH3(S10-) interphase cells. Using a Click-iT flow cytometric assay to directly measure mitotic protein synthesis, we find that most new protein synthesis during mitosis is cap-dependent, a result confirmed using the eIF4E/4G inhibitor drug 4E1RCat. For most cell lines tested, cap-dependent translation levels were generally similar between mitotic and interphase cells, and the majority of new mitotic protein synthesis was cap-dependent. These findings suggest that mitotic cap-dependent translation is generally sustained during mitosis by CDK1 phosphorylation of 4E-BP1 even under conditions of reduced mTOR signaling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos Transformantes de Poliomavirus/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Biossíntese de Proteínas , Serina-Treonina Quinases TOR/metabolismo , Proteína Quinase CDC2 , Proteínas de Ciclo Celular , Proliferação de Células , Células HEK293 , Células HeLa , Humanos , Interfase , Mitose , Nocodazol/química , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína
6.
Proc Natl Acad Sci U S A ; 111(41): E4342-9, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25271323

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV) are human DNA tumor viruses that express nuclear antigens [latency-associated nuclear antigen 1 (LANA1) and Epstein-Barr nuclear antigen 1 (EBNA1)] necessary to maintain and replicate the viral genome. We report here that both LANA1 and EBNA1 undergo highly efficient +1/-2 programmed ribosomal frameshifting to generate previously undescribed alternative reading frame (ARF) proteins in their repeat regions. EBNA1(ARF) encodes a KSHV LANA-like glutamine- and glutamic acid-rich protein, whereas KSHV LANA1(ARF) encodes a serine/arginine-like protein. Repeat sequence recoding has not been described previously for human DNA viruses. Programmed frameshifting (recoding) to generate multiple proteins from one RNA sequence can increase the coding capacity of a virus, without incurring a selective penalty against increased capsid size. The presence of similar repeat sequences in cellular genes, such as huntingtin, suggests that a comparison of repeat recoding in virus and human systems may provide functional and mechanistic insights for both systems.


Assuntos
Antígenos Virais/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Proteínas Nucleares/metabolismo , Fases de Leitura/genética , Sequências Repetitivas de Aminoácidos , Sequência de Aminoácidos , Antígenos Virais/química , Linhagem Celular Tumoral , Citoplasma/metabolismo , Antígenos Nucleares do Vírus Epstein-Barr/química , Mudança da Fase de Leitura do Gene Ribossômico , Células HEK293 , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/química , Iniciação Traducional da Cadeia Peptídica , Estrutura Terciária de Proteína
7.
J Virol ; 89(8): 4191-200, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25631078

RESUMO

UNLABELLED: Merkel cell polyomavirus (MCV) is a newly discovered human cancer virus encoding a small T (sT) oncoprotein. We performed MCV sT FLAG-affinity purification followed by mass spectroscopy (MS) analysis, which identified several protein phosphatases (PP), including PP2A A and C subunits and PP4C, as potential cellular interacting proteins. PP2A targeting is critical for the transforming properties of nonhuman polyomaviruses, such as simian virus 40 (SV40), but is not required for MCV sT-induced rodent cell transformation. We compared similarities and differences in PP2A binding between MCV and SV40 sT. While SV40 sT coimmunopurified with subunits PP2A Aα and PP2A C, MCV sT coimmunopurified with PP2A Aα, PP2A Aß, and PP2A C. Scanning alanine mutagenesis at 29 sites across the MCV sT protein revealed that PP2A-binding domains lie on the opposite molecular surface from a previously described large T stabilization domain (LSD) loop that binds E3 ligases, such as Fbw7. MCV sT-PP2A interactions can be functionally distinguished by mutagenesis from MCV sT LSD-dependent 4E-BP1 hyperphosphorylation and viral DNA replication enhancement. MCV sT has a restricted range for PP2A B subunit substitution, inhibiting only the assembly of B56α into the phosphatase holoenzyme. In contrast, SV40 sT inhibits the assembly of B55α, B56α and B56ε into PP2A. We conclude that MCV sT is required for Merkel cell carcinoma growth, but its in vitro transforming activity depends on LSD interactions rather than PP2A targeting. IMPORTANCE: Merkel cell polyomavirus is a newly discovered human cancer virus that promotes cancer, in part, through expression of its small T (sT) oncoprotein. Animal polyomavirus sT oncoproteins have been found to cause experimental tumors by blocking the activities of a group of phosphatases called protein phosphatase 2A (PP2A). Our structural analysis reveals that MCV sT also displaces the B subunit of PP2A to inhibit PP2A activity. MCV sT, however, only displaces a restricted subset of PP2A B subunits, which is insufficient to cause tumor cell formation in vitro. MCV sT instead transforms tumor cells through another region called the large T stabilization domain. The PP2A targeting and transforming activities lie on opposite faces of the MCV sT molecule and can be genetically separated from each other.


Assuntos
Antígenos Transformantes de Poliomavirus/metabolismo , Transformação Celular Neoplásica/metabolismo , Poliomavírus das Células de Merkel/metabolismo , Proteína Fosfatase 2/metabolismo , Antígenos Transformantes de Poliomavirus/genética , Cromatografia de Afinidade , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Espectrometria de Massas , Mutagênese , Ligação Proteica , Estrutura Terciária de Proteína
8.
J Virol ; 87(5): 2744-55, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23255808

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) latency associated-nuclear antigen 1 (LANA1) protein is constitutively expressed in all KSHV-infected cells, as well as in all forms of KSHV-associated malignancies. LANA1 is a multifunctional KSHV oncoprotein containing multiple repeat sequences that is important for viral episome maintenance and the regulation of cellular and viral gene expression. We characterize here multiple LANA1 isoforms and show that ∼50% of LANA1 is naturally generated as N-terminally truncated shoulder proteins that are detected on SDS-PAGE as faster-migrating shoulder bands designated LANA1(S). Higher-molecular-weight LANA1(S) isoforms initiate downstream at noncanonical sites within the N-terminal region, whereas lower-molecular-weight LANA1(S) isoforms initiate downstream within the central repeat 1 domain. LANA1(S) proteins lack an N-terminal nuclear localization signal motif, and some isoforms differ from full-length, canonical LANA1 by localizing to perinuclear and cytoplasmic sites. Although LANA1 has until now been assumed to be solely active in the nucleus, this finding indicates that this major KSHV oncoprotein may have cytoplasmic activities as well. KSHV overcomes its limited genetic coding capacity by generating alternatively initiated protein isoforms that may have distinct biological functions.


Assuntos
Antígenos Virais/química , Antígenos Virais/metabolismo , Citoplasma/metabolismo , Herpesvirus Humano 8/genética , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Iniciação Traducional da Cadeia Peptídica , Antígenos Virais/genética , Linhagem Celular , DNA Viral/genética , Células HEK293 , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Nucleares/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína
9.
J Virol ; 87(23): 13009-19, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24067965

RESUMO

Establishment of a chronic infection is a key event in virus-mediated carcinogenesis. Several cancer-associated, double-stranded DNA (dsDNA) viruses act via their oncoproteins to downregulate Toll-like receptor 9 (TLR9), a key receptor in the host innate immune response that senses viral or bacterial dsDNA. A novel oncogenic virus, Merkel cell polyomavirus (MCPyV), has been recently identified that causes up to 80% of Merkel cell carcinomas (MCCs). However, it is not yet known whether this oncogenic virus also disrupts immune-related pathways. We find that MCPyV large T antigen (LT) expression downregulates TLR9 expression in epithelial and MCC-derived cells. Accordingly, silencing of LT expression results in upregulation of mRNA TLR9 levels. In addition, small T antigen (sT) also appears to inhibit TLR9 expression, since inhibition of its expression also resulted in an increase of TLR9 mRNA levels. LT inhibits TLR9 expression by decreasing the mRNA levels of the C/EBPß transactivator, a positive regulator of the TLR9 promoter. Chromatin immunoprecipitation reveals that C/EBPß binding at a C/EBPß response element (RE) in the TLR9 promoter is strongly inhibited by expression of MCPyV early genes and that mutation of the C/EBP RE prevents MCPyV downregulation of TLR9. A survey of BK polyomavirus (BKPyV), JC polyomavirus (JCPyV), KI polyomavirus (KIPyV), MCPyV, simian virus 40 (SV40), and WU polyomavirus (WUPyV) early genes revealed that only BKPyV and MCPyV are potent inhibitors of TLR9 gene expression. MCPyV LT targeting of C/EBP transactivators is likely to play an important role in viral persistence and potentially inhibit host cell immune responses during MCPyV tumorigenesis.


Assuntos
Antígenos Virais de Tumores/metabolismo , Carcinoma de Célula de Merkel/genética , Regulação para Baixo , Poliomavírus das Células de Merkel/metabolismo , Infecções por Polyomavirus/genética , Receptor Toll-Like 9/genética , Antígenos Virais de Tumores/genética , Carcinoma de Célula de Merkel/metabolismo , Carcinoma de Célula de Merkel/virologia , Interações Hospedeiro-Patógeno , Humanos , Células de Merkel/metabolismo , Células de Merkel/virologia , Poliomavírus das Células de Merkel/genética , Infecções por Polyomavirus/metabolismo , Infecções por Polyomavirus/virologia , Regiões Promotoras Genéticas , Receptor Toll-Like 9/metabolismo , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/metabolismo , Infecções Tumorais por Vírus/virologia
10.
mBio ; : e0111724, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38940554

RESUMO

Merkel cell polyomavirus (MCPyV) is a double-stranded tumor virus that is the main causative agent of Merkel cell carcinoma (MCC). The MCPyV large T antigen (LT), an essential viral DNA replication protein, maintains viral persistence by interacting with host Skp1-Cullin 1-F-box (SCF) E3 ubiquitin ligase complexes, which subsequently induces LT's proteasomal degradation, restricting MCPyV DNA replication. SCF E3 ubiquitin ligases require their substrates to be phosphorylated to bind them, utilizing phosphorylated serine residues as docking sites. The MCPyV LT unique region (MUR) is highly phosphorylated and plays a role in multiple host protein interactions, including SCF E3 ubiquitin ligases. Therefore, this domain highly governs LT stability. Though much work has been conducted to identify host factors that restrict MCPyV LT protein expression, the kinase(s) that cooperates with the SCF E3 ligase remains unknown. Here, we demonstrate that casein kinase 1 alpha (CK1α) negatively regulates MCPyV LT stability and LT-mediated replication by modulating interactions with the SCF ß-TrCP. Specifically, we show that numerous CK1 isoforms (α, δ, ε) localize in close proximity to MCPyV LT through in situ proximity ligation assays (PLA) and CK1α overexpression mainly resulted in decreased MCPyV LT protein expression. Inhibition of CK1α using short hairpin RNA (shRNA) and treatment of a CK1α inhibitor or an mTOR inhibitor, TORKinib, resulted in decreased ß-TrCP interaction with LT, increased LT expression, and enhanced MCPyV replication. The expression level of the CSNK1A1 gene transcripts is higher in MCPyV-positive MCC, suggesting a vital role of CK1α in limiting MCPyV replication required for establishing persistent infection. IMPORTANCE: Merkel cell polyomavirus (MCPyV) large tumor antigen is a polyphosphoprotein and the phosphorylation event is required to modulate various functions of LT, including viral replication. Therefore, cellular kinase pathways are indispensable for governing MCPyV polyomavirus infection and life cycle in coordinating with the immunosuppression environment at disease onset. Understanding the regulation mechanisms of MCPyV replication by viral and cellular factors will guide proper prevention strategies with targeted inhibitors for MCPyV-associated Merkel cell carcinoma (MCC) patients, who currently lack therapies.

11.
Cells ; 12(3)2023 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-36766726

RESUMO

Senescent cells accumulate in the host during the aging process and are associated with age-related pathogeneses, including cancer. Although persistent senescence seems to contribute to many aspects of cellular pathways and homeostasis, the role of senescence in virus-induced human cancer is not well understood. Merkel cell carcinoma (MCC) is an aggressive skin cancer induced by a life-long human infection of Merkel cell polyomavirus (MCPyV). Here, we show that MCPyV large T (LT) antigen expression in human skin fibroblasts causes a novel nucleolar stress response, followed by p21-dependent senescence and senescence-associated secretory phenotypes (SASPs), which are required for MCPyV genome maintenance. Senolytic and navitoclax treatments result in decreased senescence and MCPyV genome levels, suggesting a potential therapeutic for MCC prevention. Our results uncover the mechanism of a host stress response regulating human polyomavirus genome maintenance in viral persistency, which may lead to targeted intervention for MCC.


Assuntos
Carcinoma de Célula de Merkel , Poliomavírus das Células de Merkel , Infecções por Polyomavirus , Neoplasias Cutâneas , Infecções Tumorais por Vírus , Humanos , Infecções por Polyomavirus/genética , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/patologia , Antígenos Virais de Tumores/genética , Poliomavírus das Células de Merkel/genética , Poliomavírus das Células de Merkel/metabolismo , Carcinoma de Célula de Merkel/genética , Carcinoma de Célula de Merkel/patologia , Neoplasias Cutâneas/patologia , Senescência Celular , Genoma Viral
12.
Viruses ; 14(9)2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-36146655

RESUMO

Merkel cell carcinoma (MCC) is a rare but aggressive form of skin cancer predominantly caused by the human Merkel cell polyomavirus (MCPyV). Treatment for MCC includes excision and radiotherapy of local disease, and chemotherapy or immunotherapy for metastatic disease. The schweinfurthin family of natural compounds previously displayed potent and selective growth inhibitory activity against the NCI-60 panel of human-derived cancer cell lines. Here, we investigated the impact of schweinfurthin on human MCC cell lines. Treatment with the schweinfurthin analog, 5'-methylschweinfurth G (MeSG also known as TTI-3114), impaired metabolic activity through induction of an apoptotic pathway. MeSG also selectively inhibited PI3K/AKT and MAPK/ERK pathways in the MCPyV-positive MCC cell line, MS-1. Interestingly, expression of the MCPyV small T (sT) oncogene selectively sensitizes mouse embryonic fibroblasts to MeSG. These results suggest that the schweinfurthin family of compounds display promising potential as a novel therapeutic option for virus-induced MCCs.


Assuntos
Carcinoma de Célula de Merkel , Poliomavírus das Células de Merkel , Infecções por Polyomavirus , Neoplasias Cutâneas , Infecções Tumorais por Vírus , Animais , Carcinoma de Célula de Merkel/patologia , Fibroblastos/metabolismo , Guanosina/análogos & derivados , Humanos , Poliomavírus das Células de Merkel/genética , Camundongos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Estilbenos , Tionucleosídeos
13.
Proc Natl Acad Sci U S A ; 105(42): 16272-7, 2008 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-18812503

RESUMO

Merkel cell polyomavirus (MCV) is a virus discovered in our laboratory at the University of Pittsburgh that is monoclonally integrated into the genome of approximately 80% of human Merkel cell carcinomas (MCCs). Transcript mapping was performed to show that MCV expresses transcripts in MCCs similar to large T (LT), small T (ST), and 17kT transcripts of SV40. Nine MCC tumor-derived LT genomic sequences have been examined, and all were found to harbor mutations prematurely truncating the MCV LT helicase. In contrast, four presumed episomal viruses from nontumor sources did not possess this T antigen signature mutation. Using coimmunoprecipitation and origin replication assays, we show that tumor-derived virus mutations do not affect retinoblastoma tumor suppressor protein (Rb) binding by LT but do eliminate viral DNA replication capacity. Identification of an MCC cell line (MKL-1) having monoclonal MCV integration and the signature LT mutation allowed us to functionally test both tumor-derived and wild type (WT) T antigens. Only WT LT expression activates replication of integrated MCV DNA in MKL-1 cells. Our findings suggest that MCV-positive MCC tumor cells undergo selection for LT mutations to prevent autoactivation of integrated virus replication that would be detrimental to cell survival. Because these mutations render the virus replication-incompetent, MCV is not a "passenger virus" that secondarily infects MCC tumors.


Assuntos
Antígenos Virais de Tumores/imunologia , Carcinoma de Célula de Merkel/metabolismo , Carcinoma de Célula de Merkel/virologia , Polyomavirus/fisiologia , Sequência de Aminoácidos , Antígenos Virais de Tumores/química , Antígenos Virais de Tumores/genética , Carcinoma de Célula de Merkel/genética , Carcinoma de Célula de Merkel/imunologia , Linhagem Celular Tumoral , Sequência Conservada , Humanos , Dados de Sequência Molecular , Mutação/genética , Alinhamento de Sequência , Transcrição Gênica/genética , Replicação Viral
14.
J Virol ; 83(23): 12118-28, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19759150

RESUMO

Merkel cell polyomavirus (MCV) is a recently discovered human polyomavirus causing the majority of human Merkel cell carcinomas. We mapped a 71-bp minimal MCV replication core origin sufficient for initiating eukaryotic DNA replication in the presence of wild-type MCV large T protein (LT). The origin includes a poly(T)-rich tract and eight variably oriented, GAGGC-like pentanucleotide sequences (PS) that serve as LT recognition sites. Mutation analysis shows that only four of the eight PS are required for origin replication. A single point mutation in one origin PS from a naturally occurring, tumor-derived virus reduces LT assembly on the origin and eliminates viral DNA replication. Tumor-derived LT having mutations truncating either the origin-binding domain or the helicase domain also prevent LT-origin assembly. Optimal MCV replication requires coexpression of MCV small T protein (sT), together with LT. An intact DnaJ domain on the LT is required for replication but is dispensable on the sT. In contrast, PP2A targeting by sT is required for enhanced replication. The MCV origin provides a novel model for eukaryotic replication from a defined DNA element and illustrates the selective pressure within tumors to abrogate independent MCV replication.


Assuntos
Antígenos Virais de Tumores/fisiologia , Células de Merkel/virologia , Polyomavirus/fisiologia , Origem de Replicação/genética , Replicação Viral , Sequência de Bases , Sítios de Ligação , Linhagem Celular , DNA Viral/genética , DNA Viral/metabolismo , Humanos , Dados de Sequência Molecular , Mutação Puntual , Polyomavirus/genética , Ligação Proteica
15.
Viruses ; 12(9)2020 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-32962090

RESUMO

Merkel cell polyomavirus (MCV) is the only known human oncogenic virus in the polyomaviridae family and the etiological agent of most Merkel cell carcinomas (MCC). MCC is an aggressive and highly metastatic skin cancer with a propensity for recurrence and poor prognosis. Large tumor antigen (LT), is an essential oncoprotein for MCV transcription, viral replication, and cancer cell proliferation. MCV LT is a short-lived protein that encodes a unique domain: MCV LT unique regions (MURs). These domains consist of phosphorylation sites that interact with multiple E3 ligases, thus limiting LT expression and consequently, viral replication. In this study, we show that MURs are necessary for regulating LT stability via multiple E3 ligase interactions, resulting in cell growth arrest. While expression of wild-type MCV LT induced a decrease in cellular proliferation, deletion of the MUR domains resulted in increased LT stability and cell proliferation. Conversely, addition of MURs to SV40 LT propagated E3 ligase interactions, which in turn, reduced SV40 LT stability and decreased cell growth activity. Our results demonstrate that compared to other human polyomaviruses (HPyVs), MCV LT has evolved to acquire the MUR domains that are essential for MCV LT autoregulation, potentially leading to viral latency and MCC.


Assuntos
Antígenos Virais de Tumores/metabolismo , Ciclo Celular/fisiologia , Poliomavírus das Células de Merkel/metabolismo , Estabilidade Proteica , Carcinoma de Célula de Merkel/virologia , Proliferação de Células , Células HEK293 , Humanos , Recidiva Local de Neoplasia , Polyomavirus , Infecções por Polyomavirus/virologia , Neoplasias Cutâneas/virologia , Infecções Tumorais por Vírus/virologia , Ubiquitina-Proteína Ligases , Latência Viral/fisiologia , Replicação Viral
16.
Oncogenesis ; 9(5): 53, 2020 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-32427880

RESUMO

Merkel cell polyomavirus (MCV) small T (sT) is the main oncoprotein in Merkel cell carcinoma (MCC) development. A unique domain of sT, LT stabilization domain (LSD), has been reported to bind and inactivate multiple SCF (Skp1-Cullin-F-box) E3 ligases. These interactions impede the turnover of MCV large T (LT) antigen and cellular oncoproteins such as c-Myc and cyclin E, thereby promoting viral replication and cell transformation. However, it is currently unclear how this LSD region contributes to multiple transforming activities of sT. Structural docking simulation of sT and F-box and WD repeat domain-containing 7 (FBW7) revealed a novel allosteric interaction between sT and FBW7 WD40 domain. This model is supported by experimental evidence confirming that charge engineering in the LSD alters sT-WD40 binding. Specifically, loss of net positive charge in the LSD prevents sT-FBW7 binding by abrogating the electrostatic interaction, thus impeding inhibition of FBW7 by sT. Furthermore, positively charged mutations in the LSD significantly restored the sT function and its ability to transform rodent fibroblast cells. We infer that the surface charge of sT is a major determinant for targeting E3 ligases, which leads to sT-induced cell transformation, an observation that could be used to develop targeted therapeutics for MCC.

17.
Int J Cancer ; 125(6): 1243-9, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19499546

RESUMO

Merkel cell polyomavirus (MCV) is a recently discovered human virus closely related to African green monkey lymphotropic polyomavirus. MCV DNA is integrated in approximately 80% of Merkel cell carcinomas (MCC), a neuroendocrine skin cancer linked to lymphoid malignancies such as chronic lymphocytic leukemia (CLL). To assess MCV infection and its association with human diseases, we developed a monoclonal antibody that specifically recognizes endogenous and transfected MCV large T (LT) antigen. We show expression of MCV LT protein localized to nuclei of tumor cells from MCC having PCR quantified MCV genome at an average of 5.2 (range 0.8-14.3) T antigen DNA copies per cell. Expression of this putative viral oncoprotein in tumor cells provides the mechanistic underpinning supporting the notion that MCV causes a subset of MCC. In contrast, although 2.2% of 325 hematolymphoid malignancies surveyed also showed evidence for MCV infection by DNA PCR, none were positive at high viral copy numbers, and none of 173 lymphoid malignancies examined on tissue microarrays expressed MCV LT protein in tumor cells. As with some of the other human polyomaviruses, lymphocytes may serve as a tissue reservoir for MCV infection, but hematolymphoid malignancies associated with MCC are unlikely to be caused by MCV.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Carcinoma de Célula de Merkel/virologia , Regulação Viral da Expressão Gênica , Tecido Linfoide/virologia , Linfoma/virologia , Infecções por Polyomavirus/genética , Polyomavirus/patogenicidade , Neoplasias Cutâneas/virologia , Sequência de Aminoácidos , Carcinoma de Célula de Merkel/patologia , DNA Viral/análise , Imunofluorescência , Dosagem de Genes , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Infecções por Polyomavirus/patologia , Homologia de Sequência de Aminoácidos , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/patologia
18.
Oncogene ; 24(44): 6617-25, 2005 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-16007161

RESUMO

E-cadherin is a key cell adhesion molecule implicated as a tumor suppressor, which is frequently altered in hepatocellular carcinoma, especially in hepatitis B virus (HBV)-related tumors. Here, we report that HBV X protein (HBx) represses E-cadherin expression at the transcription level. Based on the differential effects of HBx natural variants, we determined that Lys-130 in the transactivation domain of HBx is critical for the E-cadherin repression. The repression effect of HBx was abolished after treatment with DNA methyltransferase inhibitor, 5'-Aza-2'dC. In addition, methylation-specific PCR analysis revealed that the CpG island 1 of E-cadherin promoter is hypermethylated by HBx. Furthermore, HBx induces DNA methyltransferase 1 expression by stimulating its transcription. Therefore, we conclude that HBx represses E-cadherin expression by inducing methylation-mediated promoter inactivation. The reduced E-cadherin expression results in dramatic morphological changes of the HBx-expressing cells. In addition, HBx-expressing cells aggregate poorly in suspension culture, reflecting their altered intercellular interactions. The biological significance was further demonstrated by the increased collagen invasion ability of HBx-expressing cells. Therefore, the present study suggests that HBx plays a role during hepatocellular carcinogenesis by favoring cell detachment from the surrounding cells and migration outside of the primary tumor site.


Assuntos
Caderinas/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/fisiologia , Sequência de Bases , Caderinas/genética , Linhagem Celular , Metilação de DNA , Primers do DNA , Ativação Enzimática , Regulação da Expressão Gênica/fisiologia , Humanos , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais Reguladoras e Acessórias
19.
Nucleic Acids Res ; 32(7): 2202-13, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15107488

RESUMO

Despite the extensive studies on the roles of hepatitis B virus X protein (HBx), the effects of HBx on the important cellular processes such as cell growth, cell transformation and apoptosis remain controversial. Our previous study showed that the balance between p53-dependent activation and p53-independent repression by HBx determines the expression level of cyclin-dependent kinase inhibitor p21. In the present study, we further demonstrate that HBx natural variants have differential effects on p21 expression. The critical sites in HBx were identified as residues Ser-101 for activation and Met-130 for repression, respectively. The HBx variants with Ser-101 instead of Pro-101 stabilized p53 more efficiently, probably by protecting it from the MDM2-mediated degradation. On the other hand, the Met-130-containing HBx strongly repressed p21 expression by inhibiting Sp1 activity. Overall, the effect of HBx on p21 expression seems to be determined by the balance between the opposite activities. Depending on their potentials to regulate p21 expression, HBx variants showed different effects on the cell cycle progression, and eventually on the cell growth rate, implicating its biological significance. The present study may provide a clue to explaining the contradictory results related to cell growth regulation by HBx as well as to understanding the progression of hepatic diseases in HBV-positive patients.


Assuntos
Ciclinas/genética , Regulação da Expressão Gênica , Transativadores/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Carcinoma Hepatocelular/genética , Ciclo Celular , Divisão Celular , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21 , Hepatite B/genética , Humanos , Dados de Sequência Molecular , Mutação/genética , Regiões Promotoras Genéticas/genética , Transativadores/química , Transativadores/genética , Proteínas Virais Reguladoras e Acessórias
20.
FEBS Lett ; 544(1-3): 38-44, 2003 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-12782287

RESUMO

Phospholipase D (PLD) is known to stimulate cell cycle progression and to transform murine fibroblast cells into tumorigenic forms, although the precise mechanisms are not elucidated. In this report, we demonstrated that both PLD1 and PLD2 repressed expression of cyclin-dependent kinase inhibitor p21 gene in an additive manner. The phospholipase activity of PLDs was important for the effect. PLD1 repressed the p21 promoter by decreasing the level of p53, whereas PLD2 via a p53-independent pathway through modulating Sp1 activity. Taken together, we suggest that PLD isozymes stimulate cell growth by repressing expression of p21 gene, which may ultimately lead to carcinogenesis.


Assuntos
Ciclinas/metabolismo , Fosfolipase D/metabolismo , Transcrição Gênica , Sequência de Bases , Sítios de Ligação , Western Blotting , Inibidor de Quinase Dependente de Ciclina p21 , Células HeLa , Humanos , Luciferases/metabolismo , Dados de Sequência Molecular , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Homologia de Sequência do Ácido Nucleico , Fator de Transcrição Sp1/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa