Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(1): e1010753, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36689549

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS), a hyperplasia consisting of enlarged malformed vasculature and spindle-shaped cells, the main proliferative component of KS. While spindle cells express markers of lymphatic and blood endothelium, the origin of spindle cells is unknown. Endothelial precursor cells have been proposed as the source of spindle cells. We previously identified two types of circulating endothelial colony forming cells (ECFCs), ones that expressed markers of blood endothelium and ones that expressed markers of lymphatic endothelium. Here we examined both blood and lymphatic ECFCs infected with KSHV. Lymphatic ECFCs are significantly more susceptible to KSHV infection than the blood ECFCs and maintain the viral episomes during passage in culture while the blood ECFCs lose the viral episome. Only the KSHV-infected lymphatic ECFCs (K-ECFCLY) grew to small multicellular colonies in soft agar whereas the infected blood ECFCs and all uninfected ECFCs failed to proliferate. The K-ECFCLYs express high levels of SOX18, which supported the maintenance of high copy number of KSHV genomes. When implanted subcutaneously into NSG mice, the K-ECFCLYs persisted in vivo and recapitulated the phenotype of KS tumor cells with high number of viral genome copies and spindling morphology. These spindle cell hallmarks were significantly reduced when mice were treated with SOX18 inhibitor, SM4. These data suggest that KSHV-infected lymphatic ECFCs can be utilized as a KSHV infection model for in vivo translational studies to test novel inhibitors representing potential treatment modalities for KS.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Animales , Ratones , Herpesvirus Humano 8/genética , Células Endoteliales , Endotelio Vascular/patología
2.
Nucleic Acids Res ; 49(17): 10018-10033, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34417603

RESUMEN

MicroRNAs (miRNAs) are small regulatory RNAs involved in virtually all biological processes. Although many of them are co-expressed from clusters, little is known regarding the impact of this organization on the regulation of their accumulation. In this study, we set to decipher a regulatory mechanism controlling the expression of the ten clustered pre-miRNAs from Kaposi's sarcoma associated herpesvirus (KSHV). We measured in vitro the efficiency of cleavage of each individual pre-miRNA by the Microprocessor and found that pre-miR-K1 and -K3 were the most efficiently cleaved pre-miRNAs. A mutational analysis showed that, in addition to producing mature miRNAs, they are also important for the optimal expression of the whole set of miRNAs. We showed that this feature depends on the presence of a canonical pre-miRNA at this location since we could functionally replace pre-miR-K1 by a heterologous pre-miRNA. Further in vitro processing analysis suggests that the two stem-loops act in cis and that the cluster is cleaved in a sequential manner. Finally, we exploited this characteristic of the cluster to inhibit the expression of the whole set of miRNAs by targeting the pre-miR-K1 with LNA-based antisense oligonucleotides in cells either expressing a synthetic construct or latently infected with KSHV.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/genética , MicroARNs/genética , Procesamiento Postranscripcional del ARN/genética , ARN Viral/genética , Línea Celular , Células HEK293 , Humanos , Oligonucleótidos Antisentido/genética , Pliegue del ARN/genética
3.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37047272

RESUMEN

After seeing a dramatic increase in the development and use of immunotherapy and precision medicine over the past few decades, oncological care now embraces the start of the adoptive cell therapy (ACT) era. This impulse towards a new treatment paradigm has been led by chimeric antigen receptor (CAR) T cells, the only type of ACT medicinal product to be commercialized so far. Brought about by an ever-growing understanding of cellular engineering, CAR T cells are T lymphocytes genetically modified with an appropriate DNA construct, which endows them with expression of a CAR, a fusion protein between a ligand-specific recognition domain, often an antibody-like structure, and the activating signaling domain of the T cell receptor. Through this genetic enhancement, CAR T cells are engineered from a cancer patient's own lymphocytes to better target and kill their cancer cells, and the current amassed data on clinical outcomes point to a stream of bright developments in the near future. Herein, from concept design and present-day manufacturing techniques to pressing hurdles and bright discoveries around the corner, we review and thoroughly describe the state of the art in CAR T cell therapy.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Humanos , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T , Neoplasias/terapia
4.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32295923

RESUMEN

The anaphase-promoting complex, or cyclosome (APC/C), is a large E3 ubiquitin ligase composed of 14 subunits. The activity of APC/C oscillates during the cell cycle to ensure a timely transition through each phase by promoting the degradation of important cell cycle regulators. Of the human herpesviruses, cytomegalovirus (HCMV) and Epstein-Barr virus (EBV) both impair the activity of APC/C during their lytic replication cycle through virus-encoded protein kinases. Here, we addressed whether the oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) deregulates the activity of APC/C during the lytic replication cycle. To this end, we used the well-characterized iSLK.219 cell model of KSHV infection and established a new infection model of primary lymphatic endothelial cells (LECs) infected with a lytically replicating KSHV BAC16 mutant. In contrast to those of EBV and HCMV, the KSHV lytic cycle occurs while the APC/C is active. Moreover, interfering with the activity of APC/C did not lead to major changes in the production of infectious virus. We further investigated whether rereplication stress induced by the unscheduled activation of the APC/C-CDH1 complex affects the number and integrity of KSHV viral episomes. Deep sequencing of the viral episomes and host chromosomes in iSLK.219 cells revealed that, while distinct regions in the cellular chromosomes were severely affected by rereplication stress, the integrity of the viral episomes remained unaltered.IMPORTANCE DNA viruses have evolved complex strategies to gain control over the cell cycle. Several of them target APC/C, a key cellular machinery that controls the timely progression of the cell cycle, by either blocking or enhancing its activity. Here, we investigated the activity of APC/C during the lytic replication cycle of KSHV and found that, in contrast to that of KSHV's close relatives EBV and HCMV, KSHV lytic replication occurs while the APC/C is active. Perturbing APC/C activity by depleting a core protein or the adaptor proteins of the catalytic domain, and hence interfering with normal cell-cycle progression, did not affect virus replication. This suggests that KSHV has evolved to replicate independently of the activity of APC/C and in various cell cycle conditions.


Asunto(s)
Herpesvirus Humano 8/metabolismo , Latencia del Virus/genética , Replicación Viral/fisiología , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Cultivo Primario de Células , Sarcoma de Kaposi/virología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas Virales/metabolismo , Activación Viral/genética , Latencia del Virus/fisiología
5.
EMBO Rep ; 18(3): 437-450, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28174209

RESUMEN

Protein phosphatase 2A (PP2A) is a critical human tumor suppressor. Cancerous inhibitor of PP2A (CIP2A) supports the activity of several critical cancer drivers (Akt, MYC, E2F1) and promotes malignancy in most cancer types via PP2A inhibition. However, the 3D structure of CIP2A has not been solved, and it remains enigmatic how it interacts with PP2A. Here, we show by yeast two-hybrid assays, and subsequent validation experiments, that CIP2A forms homodimers. The homodimerization of CIP2A is confirmed by solving the crystal structure of an N-terminal CIP2A fragment (amino acids 1-560) at 3.0 Å resolution, and by subsequent structure-based mutational analyses of the dimerization interface. We further describe that the CIP2A dimer interacts with the PP2A subunits B56α and B56γ. CIP2A binds to the B56 proteins via a conserved N-terminal region, and dimerization promotes B56 binding. Intriguingly, inhibition of either CIP2A dimerization or B56α/γ expression destabilizes CIP2A, indicating opportunities for controlled degradation. These results provide the first structure-function analysis of the interaction of CIP2A with PP2A/B56 and have direct implications for its targeting in cancer therapy.


Asunto(s)
Autoantígenos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Oncogénicas/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Autoantígenos/química , Sitios de Unión , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/química , Modelos Moleculares , Mutación , Proteínas Oncogénicas/química , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Multimerización de Proteína , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/genética , Estabilidad Proteica , Subunidades de Proteína/metabolismo , Relación Estructura-Actividad , Proteínas Supresoras de Tumor/química
6.
PLoS Pathog ; 12(2): e1005424, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26891221

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) causes Kaposi's sarcoma and certain lymphoproliferative malignancies. Latent infection is established in the majority of tumor cells, whereas lytic replication is reactivated in a small fraction of cells, which is important for both virus spread and disease progression. A siRNA screen for novel regulators of KSHV reactivation identified the E3 ubiquitin ligase MDM2 as a negative regulator of viral reactivation. Depletion of MDM2, a repressor of p53, favored efficient activation of the viral lytic transcription program and viral reactivation. During lytic replication cells activated a p53 response, accumulated DNA damage and arrested at G2-phase. Depletion of p21, a p53 target gene, restored cell cycle progression and thereby impaired the virus reactivation cascade delaying the onset of virus replication induced cytopathic effect. Herpesviruses are known to reactivate in response to different kinds of stress, and our study now highlights the molecular events in the stressed host cell that KSHV has evolved to utilize to ensure efficient viral lytic replication.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/genética , Estrés Fisiológico/genética , Replicación Viral , Línea Celular Tumoral , Replicación del ADN , Humanos , ARN Interferente Pequeño/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Activación Viral/fisiología , Latencia del Virus/genética , Replicación Viral/genética
8.
J Infect Dis ; 211(11): 1842-51, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25492914

RESUMEN

BACKGROUND: Classic Kaposi sarcoma (cKS) is an inflammatory tumor caused by human herpesvirus 8 (HHV-8) commonly observed in elderly men of Mediterranean origin. We studied a Finnish family of 5 affected individuals in 2 generations. Except for atypical mycobacterial infection of the index case, the affected individuals did not have notable histories of infection. METHODS: We performed genome and exome sequencing and mapped shared chromosomal regions to identify genetic predisposition in the family. RESULTS: We identified 12 protein-coding candidate variants that segregated in the 3 affected cousins from whom we had samples. The affected mother of the index case was an obligatory carrier. Among the 12 candidates was a rare heterozygous substitution rs141331848 (c.1337C>T, p.Thr446Ile) in the DNA-binding domain of STAT4. The variant was not present in 242 Finnish control genomes or 180 additional regional controls. Activated T-helper cells from the HHV-8-negative variant carriers showed reduced interferon γ production, compared with age and sex matched wild-type individuals. We screened STAT4 in additional 18 familial KS cases and the variant site from 56 sporadic KS cases but detected no pathogenic mutations. CONCLUSIONS: Our data suggest that STAT4 is a potential cKS-predisposition gene, but further functional and genetic validation is needed.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Factor de Transcripción STAT4/genética , Sarcoma de Kaposi/genética , Anciano , Secuencia de Aminoácidos , Femenino , Ligamiento Genético , Genoma , Humanos , Interferón gamma/inmunología , Leucocitos Mononucleares , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Sarcoma de Kaposi/inmunología , Sarcoma de Kaposi/metabolismo , Alineación de Secuencia , Análisis de Secuencia de ADN , Linfocitos T
9.
Semin Cancer Biol ; 26: 69-77, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24486643

RESUMEN

Kaposi sarcoma (KS), a viral cancer associated to Kaposi sarcoma herpesvirus (KSHV) infection, is currently the most common tumor in men in sub-Saharan Africa. KS is an angiogenic tumor and characterized by the presence of aberrant vascular structures in the lesion. Although our understanding of how KSHV causes the aberrant differentiation of endothelial cells and the typical vascular abnormalities in KS tumors is far from complete, the experimental evidence reviewed here provides a comprehensive description of the role of KSHV in the pathogenesis of this unusual tumor. In contrast to other tumor viruses, whose interference with cellular processes relating to cell cycle, apoptosis and DNA damage may be at the heart of their oncogenic properties, KSHV may cause KS primarily by its ability to engage with the differentiation and function of endothelial cells. Although the intracellular pathways engaged by KSHV in the endothelial cells are being explored as drug targets, a better understanding of the impact of KSHV on endothelial cell differentiation and vasculogenesis is needed before the encouraging findings can form the basis for new targeted therapeutic approaches to KS.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/virología , Herpesvirus Humano 8/fisiología , Animales , Transformación Celular Neoplásica , Transformación Celular Viral , Modelos Animales de Enfermedad , Células Endoteliales/patología , Infecciones por Herpesviridae/tratamiento farmacológico , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Humanos , Ratones , Neovascularización Patológica/metabolismo , Fenotipo , Sarcoma de Kaposi/tratamiento farmacológico , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología
10.
JCI Insight ; 9(1)2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-37971882

RESUMEN

Despite strong indications that interactions between melanoma and lymphatic vessels actively promote melanoma progression, the molecular mechanisms are not yet completely understood. To characterize molecular factors of this crosstalk, we established human primary lymphatic endothelial cell (LEC) cocultures with human melanoma cell lines. Here, we show that coculture with melanoma cells induced transcriptomic changes in LECs and led to multiple changes in their function. WNT5B, a paracrine signaling molecule upregulated in melanoma cells upon LEC interaction, was found to contribute to the functional changes in LECs. Moreover, WNT5B transcription was regulated by Notch3 in melanoma cells following the coculture with LECs, and Notch3 and WNT5B were coexpressed in melanoma patient primary tumor and metastasis samples. Moreover, melanoma cells derived from LEC coculture escaped efficiently from the primary site to the proximal tumor-draining lymph nodes, which was impaired upon WNT5B depletion. This supported the role of WNT5B in promoting the metastatic potential of melanoma cells through its effects on LECs. Finally, DLL4, a Notch ligand expressed in LECs, was identified as an upstream inducer of the Notch3/WNT5B axis in melanoma. This study elucidated WNT5B as a key molecular factor mediating bidirectional crosstalk between melanoma cells and lymphatic endothelium and promoting melanoma metastasis.


Asunto(s)
Vasos Linfáticos , Melanoma , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Metástasis Linfática/patología , Vasos Linfáticos/patología , Melanoma/patología , Transducción de Señal , Proteínas Wnt/metabolismo
11.
J Virol ; 86(11): 5974-91, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22438557

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is etiologically associated with the angioproliferative Kaposi's sarcoma (KS). KSHV infection and the expression of latency-associated nuclear antigen (LANA-1) upregulates the angiogenic multifunctional 123-amino-acid, 14-kDa protein angiogenin (ANG), which is detected in KS lesions and in KSHV-associated primary effusion lymphoma (PEL) cells. ANG knockdown or the inhibition of ANG's nuclear translocation resulted in decreased LANA-1 gene expression and reduced KSHV-infected endothelial and PEL cell survival (Sadagopan et al., J. Virol. 83:3342-3364, 2009). Further studies here demonstrate that LANA-1 and ANG colocalize and coimmunoprecipitate in de novo infected endothelial cells and in latently infected PEL (BCBL-1 and BC-3) cells. LANA-1 and ANG interaction occurred in the absence of the KSHV genome and other viral proteins. In gel filtration chromatography analyses of BC-3 cell lysates, ANG coeluted with LANA-1, p53, and Mdm2 in high-molecular-weight fractions, and LANA-1, p53, and Mdm2 also coimmunoprecipitated with ANG. LANA-1, ANG, and p53 colocalized in KSHV-infected cells, and colocalization between ANG and p53 was also observed in LANA-1-negative cells. The deletion constructs of ANG suggested that the C-terminal region of amino acids 104 to 123 is involved in LANA-1 and p53 interactions. Silencing ANG or inhibiting its nuclear translocation resulted in decreased nuclear LANA-1 and ANG levels, decreased interactions between ANG-LANA-1, ANG-p53, and LANA-1-p53, the induction of p53, p21, and Bax proteins, the increased cytoplasmic localization of p53, the downregulation of Bcl-2, the increased cleavage of caspase-3, and the apoptosis of cells. No such effects were observed in KSHV-negative BJAB cells. The phosphorylation of p53 at serine 15, which is essential for p53 stabilization and for p53's apoptotic and cell cycle regulation functions, was increased in BCBL-1 cells transduced with short hairpin RNA targeting ANG. Together, these studies suggest that the antiapoptosis observed in KSHV-infected cells and the suppression of p53 functions are mediated in part by ANG, and KSHV has probably evolved to utilize angiogenin's multiple functions for the maintenance of its latency and cell survival. Thus, targeting ANG to induce the apoptosis of cells latently infected with KSHV is an attractive therapeutic strategy against KSHV infection and associated malignancies.


Asunto(s)
Antígenos Virales/metabolismo , Apoptosis , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno , Proteínas Nucleares/metabolismo , Ribonucleasa Pancreática/metabolismo , Línea Celular , Cromatografía en Gel , Células Endoteliales/virología , Humanos , Inmunoprecipitación , Mapeo de Interacción de Proteínas , Eliminación de Secuencia
12.
PLoS Pathog ; 7(12): e1002405, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22174674

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) encodes a cluster of twelve micro (mi)RNAs, which are abundantly expressed during both latent and lytic infection. Previous studies reported that KSHV is able to inhibit apoptosis during latent infection; we thus tested the involvement of viral miRNAs in this process. We found that both HEK293 epithelial cells and DG75 cells stably expressing KSHV miRNAs were protected from apoptosis. Potential cellular targets that were significantly down-regulated upon KSHV miRNAs expression were identified by microarray profiling. Among them, we validated by luciferase reporter assays, quantitative PCR and western blotting caspase 3 (Casp3), a critical factor for the control of apoptosis. Using site-directed mutagenesis, we found that three KSHV miRNAs, miR-K12-1, 3 and 4-3p, were responsible for the targeting of Casp3. Specific inhibition of these miRNAs in KSHV-infected cells resulted in increased expression levels of endogenous Casp3 and enhanced apoptosis. Altogether, our results suggest that KSHV miRNAs directly participate in the previously reported inhibition of apoptosis by the virus, and are thus likely to play a role in KSHV-induced oncogenesis.


Asunto(s)
Apoptosis/genética , Caspasa 3/biosíntesis , Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/genética , MicroARNs/genética , Northern Blotting , Western Blotting , Caspasa 3/genética , Línea Celular , Regulación hacia Abajo , Regulación Viral de la Expresión Génica/genética , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Mutagénesis Sitio-Dirigida , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Duodecim ; 129(15): 1545-51, 2013.
Artículo en Fi | MEDLINE | ID: mdl-24163972

RESUMEN

Viral infections are a common cause for the development of cancer. Most common among the cancer-inducing viruses are human papillomaviruses, which cause cervical cancer among other things. Cancer viruses aim to keep their host cells alive and simultaneously use their oncogenes to interfere with cellular phenomena such as proliferation and programmed cell death. The most effective way of preventing viral cancers is to reduce the frequency of cancer virus infections by using vaccinations and by intervening in high-risk behavior. In addition, the infections must be detected and treated in advance of cancer development.


Asunto(s)
Neoplasias/virología , Virosis/complicaciones , Virosis/prevención & control , Vacunas contra el Cáncer/farmacología , Humanos , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/farmacología
14.
Mol Cancer Ther ; 22(11): 1319-1331, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37486980

RESUMEN

Chimeric antigen receptor (CAR) T-cell immunotherapies for solid tumors face critical challenges such as heterogeneous antigen expression. We characterized stage-specific embryonic antigen-4 (SSEA-4) cell-surface glycolipid as a target for CAR T-cell therapy. SSEA-4 is mainly expressed during embryogenesis but is also found in several cancer types making it an attractive tumor-associated antigen. Anti-SSEA-4 CAR-T cells were generated and assessed preclinically in vitro and in vivo for antitumor response and safety. SSEA-4 CAR-T cells effectively eliminated SSEA-4-positive cells in all the tested cancer cell lines, whereas SSEA-4-negative cells lines were not targeted. In vivo efficacy and safety studies using NSG mice and the high-grade serous ovarian cancer cell line OVCAR4 demonstrated a remarkable and specific antitumor response at all the CAR T-cell doses used. At high T-cell doses, CAR T cell-treated mice showed signs of health deterioration after a follow-up period. However, the severity of toxicity was reduced with a delayed onset when lower CAR T-cell doses were used. Our data demonstrate the efficacy of anti-SSEA-4 CAR T-cell therapy; however, safety strategies, such as dose-limiting and/or equipping CAR-T cells with combinatorial antigen recognition should be implemented for its potential clinical translation.


Asunto(s)
Carcinoma , Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Humanos , Femenino , Animales , Ratones , Glicoesfingolípidos/metabolismo , Línea Celular Tumoral , Neoplasias Ováricas/metabolismo , Inmunoterapia Adoptiva , Linfocitos T , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
PLoS Pathog ; 6(3): e1000818, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20333249

RESUMEN

Nucleophosmin (NPM) is a multifunctional nuclear phosphoprotein and a histone chaperone implicated in chromatin organization and transcription control. Oncogenic Kaposi's sarcoma herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma, primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). In the infected host cell KSHV displays two modes of infection, the latency and productive viral replication phases, involving extensive viral DNA replication and gene expression. A sustained balance between latency and reactivation to the productive infection state is essential for viral persistence and KSHV pathogenesis. Our study demonstrates that the KSHV v-cyclin and cellular CDK6 kinase phosphorylate NPM on threonine 199 (Thr199) in de novo and naturally KSHV-infected cells and that NPM is phosphorylated to the same site in primary KS tumors. Furthermore, v-cyclin-mediated phosphorylation of NPM engages the interaction between NPM and the latency-associated nuclear antigen LANA, a KSHV-encoded repressor of viral lytic replication. Strikingly, depletion of NPM in PEL cells leads to viral reactivation, and production of new infectious virus particles. Moreover, the phosphorylation of NPM negatively correlates with the level of spontaneous viral reactivation in PEL cells. This work demonstrates that NPM is a critical regulator of KSHV latency via functional interactions with v-cyclin and LANA.


Asunto(s)
Quinasa 6 Dependiente de la Ciclina/metabolismo , Herpesvirus Humano 8/crecimiento & desarrollo , Proteínas Nucleares/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virología , Latencia del Virus/fisiología , Acetilación , Antígenos Virales/genética , Antígenos Virales/metabolismo , Línea Celular Tumoral , Herpesvirus Humano 8/genética , Humanos , Proteínas Nucleares/genética , Nucleofosmina , Fosforilación/fisiología , ARN Interferente Pequeño , Treonina/metabolismo , Replicación Viral/fisiología
16.
Cancer Cell ; 5(5): 465-75, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15144954

RESUMEN

Nucleophosmin (NPM, B23) is an abundant nucleolar phosphoprotein involved in ribosome biogenesis, and interacts with tumor suppressor proteins p53 and Rb. Here we show that NPM is a UV damage response protein that undergoes nucleoplasmic redistribution and regulates p53 and HDM2 levels and their interaction. By utilizing RNAi approaches and analyses of endogenous and ectopically expressed proteins, we demonstrate that NPM binds HDM2 and acts as a negative regulator of p53-HDM2 interaction. Viral stress, enforced by expression of Kaposi's sarcoma virus K cyclin, causes NPM redistribution, K cyclin-NPM association, and p53 stabilization by dissociation of HDM2-p53 complexes. The results demonstrate novel associations of HDM2 and K cyclin with NPM and implicate NPM as a crucial controller of p53 through inhibition of HDM2.


Asunto(s)
Nucléolo Celular/metabolismo , Ciclinas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Nucléolo Celular/efectos de la radiación , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Fibroblastos/efectos de la radiación , Glutatión Transferasa/metabolismo , Humanos , Ratones , Nucleofosmina , Osteosarcoma/metabolismo , Osteosarcoma/patología , Pruebas de Precipitina , Biosíntesis de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-mdm2 , ARN Interferente Pequeño/farmacología , Proteína SUMO-1/metabolismo , Proteína p53 Supresora de Tumor/química , Rayos Ultravioleta/efectos adversos , Dedos de Zinc
17.
Front Mol Med ; 2: 1049580, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-39086976

RESUMEN

Background: T cells equipped with chimeric antigen receptors (CAR) have shown remarkable efficacy in targeting B lineage malignancies. Improvement of the CAR structure is needed, however, with a view to developing flexibly modifiable spacers that are inert in interactions with unwanted cells. Specifically, binding to cells carrying receptors for IgG's crystallizable fragment (FcR), that recognize IgG-derived domains in CARs is to be avoided. Methods: Two novel CARs targeting the CD19 antigen where the IgG1-CH2 and -CH3 domains were replaced with Ig-like domains from signal-regulatory protein α (SIRPα) were designed in silico. An IgG1-based CAR and a CAR lacking both SIRPα and IgG1 domains were used as comparators. The phenotype and memory phenotype of the expanded cells were analyzed by flow cytometry, and CAR T cell activation and cytotoxic efficacy were assessed in co-culture experiments in response to CD19+ target cells. Unwanted interactions with FcR-expressing myeloid cells were interrogated in co-culture assays with THP-1 monocytic cells. Results: T cells carrying the novel SIRPα-based CARs enacted potent in vitro cytotoxicity against CD19 positive B-lineage leukemia cells, comparable to traditional IgG1-based CAR T cells. Co-culture of IgG1-based CAR T cells with FcR-expressing THP-1 monocytic cells led to prominent cell surface expression of CD69 on T cells together with production of Interleukin (IL)-2 and Interferon-γ, and production of IL-1ß, indicating activation of the T cells and monocytes, respectively. Longer co-culture led to killing of the monocytes. No signs of T cell nor monocyte activation were detected in co-cultures of SIRPα-based CAR T cells with THP-1 cells. Arming T cells with the SIRPα-based CARs favored differentiation towards CD4+ phenotype during expansion, while the effects on memory phenotype of the T cells were equivalent between the SIRPα- and IgG1-based CARs. In a pilot experiment, T cells modified with one of the SIRPα-based CARs showed dose dependent leukemia cell control. Conclusion: The novel SIRPα based spacers offer a suitable backbone for developing chimeric antigen receptors that evade the off-target binding to FcR while the cells retain a favorable memory phenotype and efficient cytotoxicity, establishing a promising candidate for future in vivo and clinical testing.

18.
PLoS Pathog ; 5(3): e1000324, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19266083

RESUMEN

Host signal-transduction pathways are intimately involved in the switch between latency and productive infection of herpes viruses. As with other herpes viruses, infection by Kaposi's sarcoma herpesvirus (KSHV) displays these two phases. During latency only few viral genes are expressed, while in the productive infection the virus is reactivated with initiation of extensive viral DNA replication and gene expression, resulting in production of new viral particles. Viral reactivation is crucial for KSHV pathogenesis and contributes to the progression of KS. We have recently identified Pim-1 as a kinase reactivating KSHV upon over-expression. Here we show that another Pim family kinase, Pim-3, also induces viral reactivation. We demonstrate that expression of both Pim-1 and Pim-3 is induced in response to physiological and chemical reactivation in naturally KSHV-infected cells, and we show that they are required for KSHV reactivation under these conditions. Furthermore, our data indicate that Pim-1 and Pim-3 contribute to viral reactivation by phosphorylating the KSHV latency-associated nuclear antigen (LANA) on serine residues 205 and 206. This counteracts the LANA-mediated repression of the KSHV lytic gene transcription. The identification of Pim family kinases as novel cellular regulators of the gammaherpesvirus life cycle facilitates a deeper understanding of virus-host interactions during reactivation and may represent potential novel targets for therapeutic intervention.


Asunto(s)
Antígenos Virales/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Activación Viral , Latencia del Virus , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Interferón gamma/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-pim-1/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Células Vero , Replicación Viral
19.
Methods Mol Biol ; 2265: 141-154, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33704712

RESUMEN

Three-dimensional (3D) cell culture has allowed a deeper understanding of complex pathological and physiological processes, overcoming some of the limitations of 2D cell culture on plastic and avoiding the costs and ethical issues related to experiments involving animals. Here we describe a protocol to embed single melanoma cells alone or together with primary human lymphatic endothelial cells in a 3D cross-linked matrix, to investigate the invasion and molecular crosstalk between these two cell types, respectively. After fixation and staining with antibodies and fluorescent conjugates, phenotypic changes in both cell types can be specifically analyzed by confocal microscopy.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Endoteliales/metabolismo , Melanoma/metabolismo , Esferoides Celulares/metabolismo , Línea Celular Tumoral , Células Endoteliales/citología , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Melanoma/patología , Microscopía Confocal , Invasividad Neoplásica
20.
Nat Commun ; 12(1): 512, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479235

RESUMEN

To achieve replicative immortality, cancer cells must activate telomere maintenance mechanisms to prevent telomere shortening. ~85% of cancers circumvent telomeric attrition by re-expressing telomerase, while the remaining ~15% of cancers induce alternative lengthening of telomeres (ALT), which relies on break-induced replication (BIR) and telomere recombination. Although ALT tumours were first reported over 20 years ago, the mechanism of ALT induction remains unclear and no study to date has described a cell-based model that permits the induction of ALT. Here, we demonstrate that infection with Kaposi's sarcoma herpesvirus (KSHV) induces sustained acquisition of ALT-like features in previously non-ALT cell lines. KSHV-infected cells acquire hallmarks of ALT activity that are also observed in KSHV-associated tumour biopsies. Down-regulating BIR impairs KSHV latency, suggesting that KSHV co-opts ALT for viral functionality. This study uncovers KSHV infection as a means to study telomere maintenance by ALT and reveals features of ALT in KSHV-associated tumours.


Asunto(s)
Neoplasias/genética , Homeostasis del Telómero/genética , Acortamiento del Telómero/genética , Telómero/genética , Carcinogénesis , Línea Celular , Línea Celular Tumoral , Daño del ADN , Replicación del ADN/genética , Células HeLa , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Humanos , Hibridación Fluorescente in Situ , Neoplasias/patología , Neoplasias/virología , Proteoma/genética , Proteoma/metabolismo , Telomerasa/genética , Telomerasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA