Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Med Res Rev ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38885148

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. Histology separates two main subtypes: embryonal RMS (eRMS; 60%-70%) and alveolar RMS (aRMS; 20%-30%). The aggressive aRMS carry one of two characteristic chromosomal translocations that result in the expression of a PAX3::FOXO1 or PAX7::FOXO1 fusion transcription factor; therefore, aRMS are now classified as fusion-positive (FP) RMS. Embryonal RMS have a better prognosis and are clinically indistinguishable from fusion-negative (FN) RMS. Next to histology and molecular characteristics, RMS risk groupings are now available defining low risk tumors with excellent outcomes and advanced stage disease with poor prognosis, with an overall survival of about only 20% despite intensified multimodal treatment. Therefore, development of novel effective targeted strategies to increase survival and to decrease long-term side effects is urgently needed. Recently, immunotherapies and nanomedicine have been emerging for potent and effective tumor treatments with minimal side effects, raising hopes for effective and safe cures for RMS patients. This review aims to describe the most relevant preclinical and clinical studies in immunotherapy and targeted nanomedicine performed so far in RMS and to provide an insight in future developments.

2.
Int J Mol Sci ; 24(3)2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36768928

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. The prognosis for patients with high-grade and metastatic disease is still very poor, and survivors are burdened with long-lasting side effects. Therefore, more effective and less toxic therapies are needed. Surface proteins are ideal targets for antibody-based therapies, like bispecific antibodies, antibody-drug conjugates, or chimeric antigen receptor (CAR) T-cells. Specific surface targets for RMS are scarce. Here, we performed a surfaceome profiling based on differential centrifugation enrichment of surface/membrane proteins and detection by LC-MS on six fusion-positive (FP) RMS cell lines, five fusion-negative (FN) RMS cell lines, and three RMS patient-derived xenografts (PDXs). A total of 699 proteins were detected in the three RMS groups. Ranking based on expression levels and comparison to expression in normal MRC-5 fibroblasts and myoblasts, followed by statistical analysis, highlighted known RMS targets such as FGFR4, NCAM1, and CD276/B7-H3, and revealed AGRL2, JAM3, MEGF10, GPC4, CADM2, as potential targets for immunotherapies of RMS. L1CAM expression was investigated in RMS tissues, and strong L1CAM expression was observed in more than 80% of alveolar RMS tumors, making it a practicable target for antibody-based therapies of alveolar RMS.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa , Rabdomiosarcoma , Niño , Animales , Humanos , Xenoinjertos , Rabdomiosarcoma/metabolismo , Línea Celular , Factores de Transcripción , Modelos Animales de Enfermedad , Moléculas de Adhesión de Célula Nerviosa/uso terapéutico , Línea Celular Tumoral , Antígenos B7 , Moléculas de Adhesión Celular/uso terapéutico , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo
3.
Cancer Immunol Immunother ; 71(6): 1371-1392, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34668039

RESUMEN

Endemic Burkitt lymphoma (eBL) is an aggressive B cell cancer characterized by an IgH/c-myc translocation and the harboring of Epstein-Barr virus (EBV). Evidence accumulates that CD4 + T cells might contribute to eBL pathogenesis. Here, we investigate the presence of CD4 + T cells in primary eBL tissue and their potential dichotomous impact on an EBV-infected pre-eBL cell model using ex vivo material and in vitro co-cultures. In addition, we establish a novel method to study the effect of IgH/c-myc translocation in primary B cells by employing a CRISPR/Cas9 knock-in approach to introduce and tag de novo translocation. We unprecedently document that CD4 + T cells are present in primary eBL tumor tissue. Furthermore, we demonstrate that CD4 + T cells on the one hand suppress eBL development by killing pre-eBL cells lacking IgH/c-myc translocation in vitro and on the other hand indirectly promote eBL development by inducing crucial EBV Latency III to Latency I switching in pre-eBL cells. Finally, we show that while the mere presence of an IgH/c-myc translocation does not suffice to escape CD4 + T-cell-mediated killing in vitro, the CD4 + T-cell-mediated suppression of EBV's Latency III program in vivo may allow cells harboring an IgH/c-myc translocation and additional mutations to evade immune control and proliferate by means of deregulated c-myc activity, resulting in neoplasia. Thus, our study highlights the dichotomous effects of CD4 + T cells and the mechanisms involved in eBL pathogenesis, suggests mechanisms of their impact on eBL progression, and provides a novel in vitro model for further investigation of IgH/c-myc translocation.


Asunto(s)
Linfoma de Burkitt , Infecciones por Virus de Epstein-Barr , Linfocitos B/metabolismo , Linfoma de Burkitt/genética , Supervivencia Celular , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4 , Humanos
4.
Lab Invest ; 99(5): 612-624, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30664711

RESUMEN

Nasal natural killer/T-cell lymphoma (NNKTL) is closely associated with Epstein-Barr virus (EBV) and is characterized by poor prognosis, resulting from rapid progression of lesions in the affected organs. Recent data have shown that NNKTL is associated with the aberrant expression of cyclin-dependent kinase 1 (CDK1) and its downstream target survivin, but little is known about the functional roles of CDK1 and survivin in NNKTL. In the current study, we show that knockdown of the EBV-encoded oncoprotein latent membrane protein 1 (LMP1) induces downregulation of CDK1 and survivin in NNKTL cells. Immunohistochemistry detected CDK1 and survivin expression in LMP1-positive cells of NNKTL biopsy specimens. Inhibition of CDK1 and survivin in NNKTL cells with several inhibitors led to a dose-dependent decrease in cell proliferation. In addition, the Sp1 inhibitor mithramycin, which can downregulate both CDK1 and survivin, significantly suppressed the growth of established NNKTL in a murine xenograft model. Our results suggest that LMP1 upregulation of CDK1 and survivin may be essential for NNKTL progression. Furthermore, targeting CDK1 and survivin with Sp1 inhibitors such as mithramycin may be an effective approach to treat NNKTL, which is considered to be a treatment-refractory lymphoma.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Células Asesinas Naturales/metabolismo , Linfoma de Células T/metabolismo , Neoplasias Nasales/metabolismo , Survivin/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/genética , Línea Celular Tumoral , Femenino , Humanos , Células Asesinas Naturales/efectos de los fármacos , Linfoma de Células T/tratamiento farmacológico , Linfoma de Células T/genética , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Neoplasias Nasales/tratamiento farmacológico , Neoplasias Nasales/genética , Plicamicina/administración & dosificación , Interferencia de ARN , Survivin/antagonistas & inhibidores , Survivin/genética , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
5.
Int J Cancer ; 141(4): 654-663, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28369813

RESUMEN

Proprotein convertases are proteases that have been implicated in the activation of a wide variety of proteins. These proteins are generally synthesised as precursor proteins and require limited proteolysis for conversion into their mature bioactive counterparts. Many of these proteins, including metalloproteases, growth factors and their receptors or adhesion molecules, have been shown to facilitate tumour formation and progression. Hence, this review will focus on the proprotein convertase furin and its role in cancer. The expression of furin has been confirmed in a large spectrum of cancers such as head and neck squamous cell carcinoma, breast cancer and rhabdomyosarcoma. Functional studies modulating furin activity uncovered its importance for the processing of many cancer-related substrates and strongly indicate that high furin activity promotes the malignant phenotype of cancer cells. In this review, we summarise the expression and function of furin in different cancer types, discuss its role in processing cancer-related proproteins and give examples of potential therapeutic approaches that take advantage of the proteolytic activity of furin in cancer cells.


Asunto(s)
Furina/metabolismo , Neoplasias/metabolismo , Animales , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia
6.
J Virol ; 88(17): 10002-12, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24942583

RESUMEN

UNLABELLED: In order to understand and possibly treat B-cell malignancies associated with latent gammaherpesvirus infection, it is vital to understand the factors that control the balance between the two transcriptional states of gammaherpesviruses: latency and lytic replication. We used murine gammaherpesvirus 68 (MHV 68) as a model system to investigate how engagement of endosomal Toll-like receptors (TLRs) impacts reactivation from latency in vitro and establishment of latent infection in vivo. We found that treatment with TLR7 ligand R848 or TLR9 ligand CpG oligodeoxynucleotide (ODN) suppresses reactivation of MHV 68 in vitro. These suppressive effects correlated with the ability to activate cellular transcription factor NF-κB. Downregulation of TLR9 by RNA interference in vitro led to a reduction of nuclear levels of NF-κB p65 and consequently to an increase of spontaneous reactivation in cells latently infected with MHV 68, indicating that the TLR9 pathway suppresses spontaneous reactivation events. In vivo, sustained stimulation of TLR7 by repeated R848 treatment led to an increased frequency of infected splenocytes compared to mock-treated control results. Frequencies of infected splenic B cells in tlr7-/- or tlr9-/- mice after establishment of latency did not differ from those seen with their wild-type counterparts. Nevertheless, MHV 68-infected B cells from tlr9-/- mice showed a higher frequency of reactivation than B cells from wild-type or tlr7-/- mice in ex vivo reactivation assays. Thus, we show a suppressive effect of TLR7 or TLR9 triggering on MHV 68 reactivation that correlates with NF-κB activation and that the mere presence of a functional TLR9 signaling pathway contributes to dampen lytic gammaherpesvirus reactivation in infected cells. IMPORTANCE: A hallmark of gammaherpesviruses is their establishment of latency in B cells that is reversible through lytic reactivation. Latency can result in B-cell malignancies. Activation of the innate immune system is thought to contribute to controlling the switch between the transcriptional states of latency and reactivation. Nevertheless, the mechanisms involved are not clear. Here, we show that engagement of Toll-like receptor 7 (TLR7) and TLR9 suppresses reactivation of murine gammaherpesvirus MHV 68 in vitro and that stimulation of TLR7 in vivo increases the frequency of infected cells. TLR7 and TLR9 are innate immunity sensors of nucleic acids localized in endosomes. Additionally, we demonstrate that impairment of TLR9 signaling in latently infected B cells leads to increased reactivation. Thus, activated endosomal TLR7 and TLR9 pathways play an important role in promoting establishment of latent gammaherpesvirus infection. Counteracting signaling of these pathways allows reactivation and could represent treatment targets in gammaherpesvirus-associated malignancies.


Asunto(s)
Glicoproteínas de Membrana/inmunología , FN-kappa B/inmunología , Rhadinovirus/inmunología , Rhadinovirus/fisiología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Activación Viral , Animales , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Ratones Endogámicos C57BL , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/virología , Latencia del Virus
7.
J Infect Dis ; 209(2): 255-64, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23935199

RESUMEN

Epstein-Barr virus (EBV) infects >90% of the human population within the first 2 decades of life and establishes reversible latent infection in B cells. The stimuli that lead to switching from latent to lytic EBV infection in vivo are still elusive. Group A streptococci (GAS) are a common cause of bacterial pharyngotonsillitis in children and adolescents and colonize the tonsils and pharynx of up to 20% of healthy children. Thus, concomitant presence of EBV and GAS in the same individual is frequent. Here, we show that EBV carriers who are colonized with GAS shed EBV particles in higher numbers in their saliva, compared with EBV carriers not colonized with GAS. Messenger RNA levels of the master lytic regulatory EBV gene BZLF1 were more frequently detected in tonsils from EBV carriers colonized with GAS than from EBV carriers not colonized. Heat-killed GAS, potentially mimicking GAS colonization, elicited lytic EBV in latently infected lymphoblastoid cell lines (LCLs) partially via Toll-like receptor 2 triggering, as did purified GAS peptidoglycan. Thus, colonization by GAS might benefit EBV by increasing the EBV load in saliva and thereby enhancing the likelihood of EBV spread to other hosts.


Asunto(s)
Portador Sano/virología , Coinfección/microbiología , Coinfección/virología , Infecciones por Virus de Epstein-Barr/virología , Orofaringe/microbiología , Infecciones Estreptocócicas/microbiología , Latencia del Virus , Adolescente , Niño , Preescolar , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Herpesvirus Humano 4/aislamiento & purificación , Herpesvirus Humano 4/fisiología , Humanos , Masculino , Interacciones Microbianas , Tonsila Palatina/microbiología , ARN Mensajero/análisis , ARN Mensajero/genética , Saliva/virología , Infecciones Estreptocócicas/complicaciones , Streptococcus pyogenes/aislamiento & purificación , Streptococcus pyogenes/fisiología , Transactivadores/análisis , Transactivadores/genética , Esparcimiento de Virus
8.
Eur J Pharm Biopharm ; 194: 49-61, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029941

RESUMEN

Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. More effective and less toxic therapies are urgently needed for high-risk patients. Peptide-guided targeted drug delivery can increase the therapeutic index of encapsulated drugs and improve patients' well-being. To apply this strategy to RMS, we identified the peptide F3 in a screening for peptides binding to RMS cells surface. F3 binds to nucleolin, which is present on the surface of RMS cells and is abundantly expressed at the mRNA level in RMS patients' biopsies compared to healthy tissues. We developed a rapid microfluidic formulation of F3-decorated PEGylated liposomes and remote loading of the chemotherapeutic drug vincristine. Size, surface charge, drug loading and retention of targeted and control liposomes were studied. Enhanced cellular binding and uptake were observed in three different nucleolin-positive RMS cell lines. Importantly, F3-functionalized liposomes loaded with vincristine were up to 11 times more cytotoxic than non-targeted liposomes for RMS cell lines. These results demonstrate that F3-functionalized liposomes are promising for targeted drug delivery to RMS and warrant further in vivo investigations.


Asunto(s)
Liposomas , Rabdomiosarcoma , Niño , Humanos , Liposomas/metabolismo , Nucleolina , Vincristina/uso terapéutico , Línea Celular Tumoral , Péptidos/metabolismo , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/metabolismo
9.
Int J Cancer ; 133(10): 2341-50, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23640782

RESUMEN

Lymphoproliferative diseases (LPDs) associated with Epstein-Barr virus (EBV) infection cause significant morbidity and mortality in bone marrow and solid organ transplant recipients. To gain insight into LPD pathogenesis and to identify potential effective therapeutic approaches, we investigated early molecular events leading to B-cell transformation by gene expression profiling of EBV-infected B-cells from tonsils by Affymetrix microarray 72 hr postinfection when the B-cells hyperproliferation phase starts. Cell cycle and apoptosis were the most significantly affected pathways and enriched gene sets. In particular, we found significantly increased expression of cyclin-dependent kinase (CDK)1 and CCNB1 (cyclin B1) and of one of their downstream targets BIRC5 (survivin). Importantly, the strong upregulation of the antiapoptotic protein survivin was confirmed in lymphoblastoid cell lines (LCLs) and 71% of EBV-positive post-transplant EBV-LPD lesions scored positive for survivin. The validity of early transforming events for the identification of therapeutic targets for EBV-LPD was confirmed by the marked antiproliferative effect of the CDK inhibitor flavopiridol on LCLs and by the strong induction of apoptosis by survivin inhibition with YM155 or terameprocol. Our results suggest that targeting of CDKs and/or survivin in post-transplant EBV-LPD by specific inhibitors might be an important approach to control and eliminate EBV-transformed B-cells that should be further considered.


Asunto(s)
Linfocitos B/metabolismo , Linfocitos B/virología , Proteína Quinasa CDC2/genética , Infecciones por Virus de Epstein-Barr/genética , Proteínas Inhibidoras de la Apoptosis/genética , Trastornos Linfoproliferativos/genética , Trasplante de Órganos/efectos adversos , Apoptosis/genética , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/genética , Ciclina B1/genética , Ciclina B1/metabolismo , Infecciones por Virus de Epstein-Barr/etiología , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología , Expresión Génica , Herpesvirus Humano 4 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Trastornos Linfoproliferativos/etiología , Trastornos Linfoproliferativos/metabolismo , Survivin , Transformación Genética , Regulación hacia Arriba
10.
J Exp Clin Cancer Res ; 42(1): 293, 2023 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-37924157

RESUMEN

BACKGROUND: Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in childhood, whose prognosis is still poor especially for metastatic, high-grade, and relapsed RMS. New treatments are urgently needed, especially systemic therapies. Chimeric Antigen Receptor T cells (CAR Ts) are very effective against hematological malignancies, but their efficacy against solid tumors needs to be improved. CD276 (B7-H3) is a target upregulated in RMS and detected at low levels in normal tissues. FGFR4 is a very specific target for RMS. Here, we optimized CAR Ts for these two targets, alone or in combination, and tested their anti-tumor activity in vitro and in vivo. METHODS: Four different single-domain antibodies were used to select the most specific FGFR4-CAR construct. RMS cell killing and cytokine production by CD276- and FGFR4-CAR Ts expressing CD8α or CD28 HD/TM domains in combination with 4-1BB and/or CD28 co-stimulatory domains were tested in vitro. The most effective CD276- and FGFR4-CAR Ts were used to generate Dual-CAR Ts. Tumor killing was evaluated in vivo in three orthotopic RMS mouse models. RESULTS: CD276.V-CAR Ts (276.MG.CD28HD/TM.CD28CSD.3ζ) showed the strongest killing of RMS cells, and the highest release of IFN-γ and Granzyme B in vitro. FGFR4.V-CAR Ts (F8-FR4.CD28HD/TM.CD28CSD.3ζ) showed the most specific killing. CD276-CAR Ts successfully eradicated RD- and Rh4-derived RMS tumors in vivo, achieving complete remission in 3/5 and 5/5 mice, respectively. In CD276low JR-tumors, however, they achieved complete remission in only 1/5 mice. FGFR4 CAR Ts instead delayed Rh4 tumor growth. Dual-CAR Ts promoted Rh4-tumors clearance in 5/5 mice. CONCLUSIONS: CD276- and CD276/FGFR4-directed CAR Ts showed effective RMS cell killing in vitro and eradication of CD276high RMS tumors in vivo. CD276low tumors escaped the therapy highlighting a correlation between antigen density and effectiveness. FGFR4-CAR Ts showed specific killing in vitro but could only delay RMS growth in vivo. Our results demonstrate that combined expression of CD276-CAR with other CAR does not reduce its benefit. Introducing immunotherapy with CD276-CAR Ts in RMS seems to be feasible and promising, although CAR constructs design and target combinations have to be further improved to eradicate tumors with low target expression.


Asunto(s)
Antígenos B7 , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos , Rabdomiosarcoma , Linfocitos T , Animales , Ratones , Antígenos B7/metabolismo , Antígenos CD28/metabolismo , Línea Celular Tumoral , Recurrencia Local de Neoplasia/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Rabdomiosarcoma/terapia , Rabdomiosarcoma/patología
11.
Cancer Cell ; 4(5): 393-403, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14667506

RESUMEN

The vasculature in the angiogenic stages of a mouse model of pancreatic islet carcinogenesis was profiled in vivo with phage libraries that display short peptides. We characterized seven peptides distinguished by their differential homing to angiogenic progenitors, solid tumors, or both. None homed appreciably to normal pancreatic islets or other organs. Five peptides selectively homed to neoplastic lesions in the pancreas and not to islet beta cell tumors growing subcutaneously, xenotransplant tumors from a human cancer cell line, or an endogenously arising squamous cell tumor of the skin. Three peptides with distinctive homing to angiogenic islets, tumors, or both colocalized with markers that identify endothelial cells or pericytes. One peptide is homologous with pro-PDGF-B, which is expressed in endothelial cells, while its receptor is expressed in pericytes.


Asunto(s)
Biomarcadores de Tumor , Vasos Sanguíneos/fisiopatología , Islotes Pancreáticos/fisiopatología , Neovascularización Patológica/metabolismo , Biblioteca de Péptidos , Péptidos/metabolismo , Animales , Células Endoteliales/metabolismo , Inmunohistoquímica , Insulinoma/diagnóstico , Insulinoma/metabolismo , Ratones , Estadificación de Neoplasias , Neovascularización Patológica/diagnóstico , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/metabolismo , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo
12.
Cancers (Basel) ; 14(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36291832

RESUMEN

Active drug delivery by tumor-targeting peptides is a promising approach to improve existing therapies for rhabdomyosarcoma (RMS), by increasing the therapeutic effect and decreasing the systemic toxicity, e.g., by drug-loaded peptide-targeted nanoparticles. Here, we tested 20 different tumor-targeting peptides for their ability to bind to two RMS cell lines, Rh30 and RD, using quantum dots Streptavidin and biotin-peptides conjugates as a model for nanoparticles. Four peptides revealed a very strong binding to RMS cells: NCAM-1-targeting NTP peptide, nucleolin-targeting F3 peptide, and two Furin-targeting peptides, TmR and shTmR. F3 peptide showed the strongest binding to all RMS cell lines tested, low binding to normal control myoblasts and fibroblasts, and efficient internalization into RMS cells demonstrated by the cytoplasmic delivery of the Saporin toxin. The expression of the nucleophosphoprotein nucleolin, the target of F3, on the surface of RMS cell lines was validated by competition with the natural ligand lactoferrin, by colocalization with the nucleolin-binding aptamer AS1411, and by the marked sensitivity of RMS cell lines to the growth inhibitory nucleolin-binding N6L pseudopeptide. Taken together, our results indicate that nucleolin-targeting by F3 peptide represents a potential therapeutic approach for RMS.

13.
J Virol ; 84(13): 6667-77, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20427540

RESUMEN

Epstein-Barr virus (EBV) uses nasal mucosa-associated lymphoid tissue (NALT) as a portal of entry to establish life-long persistence in memory B cells. We previously showed that naïve and memory B cells from NALT are equally susceptible to EBV infection. Here we show that memory B cells from NALT are significantly more susceptible to EBV infection than those from remote lymphatic organs. We identify beta(1) integrin, which is expressed the most by naïve B cells of distinct lymphoid origin and by memory B cells from NALT, as a mediator of increased susceptibility to infection by EBV. Furthermore, we show that BMRF-2-beta(1) integrin interaction and the downstream signal transduction pathway are critical for postbinding events. An increase of beta(1) integrin expression in peripheral blood memory B cells provoked by CD40 stimulation plus B-cell receptor cross-linking increased the susceptibility of non-NALT memory B cells to EBV infection. Thus, EBV seems to utilize the increased activation status of memory B cells residing in the NALT to establish and ensure persistence.


Asunto(s)
Linfocitos B/virología , Herpesvirus Humano 4/crecimiento & desarrollo , Integrina beta1/biosíntesis , Línea Celular , Humanos , Glicoproteínas de Membrana/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Transducción de Señal , Proteínas Virales/metabolismo
14.
Cancers (Basel) ; 12(11)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182650

RESUMEN

The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in rhabdomyosarcoma (RMS) and represents a promising target for treatments based on specific and efficient antibodies. Despite progress, there is an urgent need for targeted treatment options to improve survival rates, and to limit long-term side effects. From phage display libraries we selected FGFR4-specific single-domain antibodies (sdAb) binding to recombinant FGFR4 and validated them by flow cytometry, surface plasmon resonance, and fluorescence microscopy. The specificity of the selected sdAb was verified on FGFR4-wild type and FGFR4-knock out cells. FGFR4-sdAb were used to decorate vincristine-loaded liposomes and to generate chimeric antigen receptor (CAR) T cells. First, incubation of RMS cells with FGFR4-sdAb revealed that FGFR4-sdAb can block FGF19-FGFR4 signaling via the MAPK pathway and could therefore serve as therapeutics for FGFR4-dependent cancers. Second, FGFR4-targeted vincristine-loaded liposomes bound specifically to RMS cells and were internalized by the receptor, demonstrating the potential for active drug delivery to the tumor. Third, FGFR4-CAR T cells, generated with one sdAb candidate, demonstrated strong and specific cytotoxicity against FGFR4 expressing RMS cells. We selected novel FGFR4-sdAb with high specificity and nano- to picomolar affinities for FGFR4 which have the potential to enable multiple FGFR4-targeted cancer therapy approaches.

15.
Biomater Sci ; 8(13): 3678-3684, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32469353

RESUMEN

Porphyrin-based periodic mesoporous organosilica nanoparticles (PMO) synthesized from a large functional octatriethoxysilylated porphyrin precursor and allowing two-photon excitation photodynamic therapy (TPE-PDT) and NIR imaging were synthesized. These PMO were grafted with polyethylene glycol (PEG) moieties and an analogue of mannose 6-phosphate functionalized at the anomeric position (AMFA). AMFAs are known to efficiently target mannose 6-phosphate receptors (M6PRs) which are over-expressed in various cancers. Here, we demonstrated for the first time that M6PRs were over-expressed in rhabdomyosarcoma (RMS) cells and could be efficiently targeted with PMO-AMFA allowing TPE imaging and TPE-PDT of RMS cells. The comparison with healthy myoblasts demonstrated an absence of biological effects, suggesting a cancer cell specificity in the biomedical action observed.


Asunto(s)
Antineoplásicos/farmacología , Materiales Biocompatibles/farmacología , Compuestos de Organosilicio/farmacología , Receptor IGF Tipo 2/antagonistas & inhibidores , Rabdomiosarcoma/tratamiento farmacológico , Nanomedicina Teranóstica , Antineoplásicos/síntesis química , Antineoplásicos/química , Materiales Biocompatibles/síntesis química , Materiales Biocompatibles/química , Humanos , Nanopartículas/química , Imagen Óptica , Compuestos de Organosilicio/síntesis química , Compuestos de Organosilicio/química , Tamaño de la Partícula , Fotoquimioterapia , Porosidad , Porfirinas/química , Porfirinas/farmacología , Proteómica , Receptor IGF Tipo 2/genética , Rabdomiosarcoma/diagnóstico por imagen , Rabdomiosarcoma/genética , Propiedades de Superficie , Células Tumorales Cultivadas
16.
Trends Microbiol ; 16(11): 520-7, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18835714

RESUMEN

The oncogenic Epstein-Barr virus (EBV) infects the majority of the human population without doing harm and establishes a latent infection in the memory B-cell compartment. To accomplish this, EBV hijacks B-cell differentiation pathways and uses its own viral genes to interfere with B-cell signalling to achieve life-long persistence. EBV latent membrane protein 2A (LMP2A) provides a surrogate B-cell receptor signal essential for cell survival and is believed to have a crucial role in the maintenance of latency by blocking B-cell activation which would otherwise lead to lytic EBV infection. These two functions demand tight control of LMP2A activity and expression levels. Based on recent insights in the function of LMP2B, an isoform of LMP2A, we propose a model for how LMP2B modulates the activity of LMP2A contributing to maintenance of EBV latency.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Proteínas de la Matriz Viral/metabolismo , Latencia del Virus , Animales , Linfocitos B/fisiología , Linfocitos B/virología , Diferenciación Celular , Herpesvirus Humano 4/genética , Humanos , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de la Matriz Viral/genética
17.
Immunology ; 128(4): 573-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19950420

RESUMEN

Toll-like receptors (TLRs) are key receptors of the innate immune system and show cell subset-specific expression. We investigated the messenger RNA (mRNA) expression of TLR genes in human haematopoietic stem cells (HSC), in naïve B cells, in memory B cells, in plasma cells from palatine tonsils and in plasma cells from peripheral blood. HSC and plasma cells showed unrestricted expression of TLR1-TLR9, in contrast to B cells which lacked TLR3, TLR4 and TLR8 but expressed mRNA of all other TLRs. We demonstrated, for the first time, that TLR triggering of terminally differentiated plasma cells augments immunoglobulin production. Thus, boosting the immediate antibody response by plasma cells upon pathogen recognition may point to a novel role of TLRs.


Asunto(s)
Inmunoglobulinas/biosíntesis , Células Plasmáticas/inmunología , Receptores Toll-Like/biosíntesis , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Regulación de la Expresión Génica/inmunología , Células Madre Hematopoyéticas/inmunología , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis , Memoria Inmunológica/inmunología , ARN Mensajero/genética , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
18.
Int J Cancer ; 124(9): 2026-32, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19123480

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. To improve existing therapies and broaden the spectrum of cytotoxic agents that can be used in RMS treatment, we performed a phage-display-based screening for peptides that bind specifically to RMS cells. Two peptides binding to RMS and to other tumour cell lines, but not to normal skeletal muscle cells and fibroblasts, were isolated from phage-displayed random peptide libraries. One peptide, named RMS-I (CQQSNRGDRKRC) contained the integrin-binding motif RGD and its binding was blocked by an antibody against alpha(v)beta(3)integrin, which is expressed on the RMS cell line RD. The isolation of RMS-I confirmed the validity of our screening procedure. The second peptide, named RMS-II (CMGNKRSAKRPC), shows sequence similarity to a previously identified peptide with tumour lymphatic specificity, LyP-1. However, RMS-II binds in vivo to RMS xenografts better than LyP-1 and homes to the tumour blood and not to lymphatic vessels. Therefore, RMS-II represents a promising peptide for the development of RMS-specific targeting approaches.


Asunto(s)
Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Biblioteca de Péptidos , Péptidos Cíclicos/química , Rabdomiosarcoma/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Fibrosarcoma/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Sistema Linfático/patología , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Neuroblastoma/metabolismo , Oligopéptidos/metabolismo , Fragmentos de Péptidos/genética , Trasplante Heterólogo
19.
J Virol ; 82(4): 1739-47, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18057232

RESUMEN

The B-lymphotropic Epstein-Barr virus (EBV) encodes two isoforms of latent membrane protein 2 (LMP2), LMP2A and LMP2B, which are expressed during latency in B cells. The function of LMP2B is largely unknown, whereas LMP2A blocks B-cell receptor (BCR) signaling transduction and induction of lytic EBV infection, thereby promoting B-cell survival. Transfection experiments on LMP2B in EBV-negative B cells and the silencing of LMP2B in EBV-harboring Burkitt's lymphoma-derived Akata cells suggest that LMP2B interferes with the function of LMP2A, but the role of LMP2B in the presence of functional EBV has not been established. Here, LMP2B, LMP2A, or both were overexpressed in EBV-harboring Akata cells to study the function of LMP2B. The overexpression of LMP2B increased the magnitude of EBV switching from its latent to its lytic form upon BCR cross-linking, as indicated by a more-enhanced upregulation and expression of EBV lytic genes and significantly increased production of transforming EBV compared to Akata vector control cells or LMP2A-overexpressing cells. Moreover, LMP2B lowered the degree of BCR cross-linking required to induce lytic EBV infection. Finally, LMP2B colocalized with LMP2A as demonstrated by immunoprecipitation and immunofluorescence and restored calcium mobilization upon BCR cross-linking, a signaling process inhibited by LMP2A. Thus, our findings suggest that LMP2B negatively regulates the function of LMP2A in preventing the switch from latent to lytic EBV replication.


Asunto(s)
Herpesvirus Humano 4/fisiología , Proteínas de la Matriz Viral/metabolismo , Activación Viral , Calcio/metabolismo , Línea Celular Tumoral , Humanos , Proteínas de la Matriz Viral/genética , Latencia del Virus
20.
J Virol ; 82(9): 4400-12, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18321980

RESUMEN

Epstein-Barr virus (EBV) uses tonsils as the portal of entry to establish persistent infection. EBV is found in various B-cell subsets in tonsils but exclusively in memory B cells in peripheral blood. The in vitro susceptibilities of B-cell subsets to EBV infection have been studied solely qualitatively. In this work, we examined quantitatively the in vitro susceptibilities of various B-cell subsets from different tissue origins to EBV infection. First, we established a centrifugation-based inoculation protocol (spinoculation) that resulted in a significantly increased proportion of infected cells compared to that obtained by conventional inoculation, enabling a detailed susceptibility analysis. Importantly, B-cell infection occurred via the known EBV receptors and infected cells showed EBV mRNA expression patterns similar to those observed after conventional inoculation, validating our approach. Tonsillar naïve and memory B cells were infected ex vivo at similar frequencies. In contrast, memory B cells from blood, which represent B cells from various lymphoid tissues, were infected at lower frequencies than their naïve counterparts. Immunoglobulin A (IgA)-positive or IgG-positive tonsillar memory B cells were significantly more susceptible to EBV infection than IgM-positive counterparts. Memory B cells were transformed with lower efficiency than naïve B cells. This result was paralleled by lower proliferation rates. In summary, these data suggest that EBV exploits the B-cell differentiation status and tissue origin to establish persistent infection.


Asunto(s)
Subgrupos de Linfocitos B/virología , Diferenciación Celular , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/virología , Sangre , Células Cultivadas , Susceptibilidad a Enfermedades/virología , Infecciones por Virus de Epstein-Barr/patología , Humanos , Memoria Inmunológica , Tejido Linfoide
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA