Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Biol Chem ; 294(44): 16297-16308, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31519750

RESUMEN

Herpesviruses can rewire cellular signaling in host cells by expressing viral G protein-coupled receptors (GPCRs). These viral receptors exhibit homology to human chemokine receptors, but some display constitutive activity and promiscuous G protein coupling. Human cytomegalovirus (HCMV) has been detected in multiple cancers, including glioblastoma, and its genome encodes four GPCRs. One of these receptors, US28, is expressed in glioblastoma and possesses constitutive activity and oncomodulatory properties. UL33, another HCMV-encoded GPCR, also displays constitutive signaling via Gαq, Gαi, and Gαs proteins. However, little is known about the nature and functional effects of UL33-driven signaling. Here, we assessed UL33's signaling repertoire and oncomodulatory potential. UL33 activated multiple proliferative, angiogenic, and inflammatory signaling pathways in HEK293T and U251 glioblastoma cells. Notably, upon infection, UL33 contributed to HCMV-mediated STAT3 activation. Moreover, UL33 increased spheroid growth in vitro and accelerated tumor growth in different in vivo tumor models, including an orthotopic glioblastoma xenograft model. UL33-mediated signaling was similar to that stimulated by US28; however, UL33-induced tumor growth was delayed. Additionally, the spatiotemporal expression of the two receptors only partially overlapped in HCMV-infected glioblastoma cells. In conclusion, our results unveil that UL33 has broad signaling capacity and provide mechanistic insight into its functional effects. UL33, like US28, exhibits oncomodulatory properties, elicited via constitutive activation of multiple signaling pathways. UL33 and US28 might contribute to HCMV's oncomodulatory effects through complementing and converging cellular signaling, and hence UL33 may represent a promising drug target in HCMV-associated malignancies.


Asunto(s)
Receptores de Quimiocina/metabolismo , Proteínas Virales/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Citomegalovirus/metabolismo , Proteínas de Unión al GTP/metabolismo , Glioblastoma/patología , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Receptores de Quimiocina/genética , Receptores Virales/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
2.
Invest New Drugs ; 36(6): 1006-1015, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29728897

RESUMEN

Introduction MCLA-128 is a bispecific monoclonal antibody targeting the HER2 and HER3 receptors. Pharmacokinetics (PK) and pharmacodynamics (PD) of MCLA-128 have been evaluated in preclinical studies in cynomolgus monkeys and mice. The aim of this study was to characterize the PK and PD of MCLA-128 and to predict a safe starting dose and efficacious clinical dose for the First-In-Human study. Methods A PK-PD model was developed based on PK data from cynomolgus monkeys and tumor growth data from a mouse JIMT-1 xenograft model. Allometric scaling was used to scale PK parameters between species. Simulations were performed to predict the safe and efficacious clinical dose, based on AUCs, receptor occupancies and PK-PD model simulations. Results MCLA-128 PK in cynomolgus monkeys was described by a two-compartment model with parallel linear and nonlinear clearance. The xenograft tumor growth model consisted of a tumor compartment with a zero-order growth rate and a first-order dying rate, both affected by MCLA-128. Human doses of 10 to 480 mg q3wk were predicted to show a safety margin of >10-fold compared to the cynomolgus monkey AUC at the no-observed-adverse-effect-level (NOAEL). Doses of ≥360 mg resulted in predicted receptor occupancies above 99% (Cmax and Cave). These doses showed anti-tumor efficacy in the PK-PD model. Conclusions This analysis predicts that a flat dose of 10 to 480 mg q3wk is suitable as starting dose for a First-in-Human study with MCLA-128. Flat doses ≥360 mg q3wk are expected to be efficacious in human, based on receptor occupancies and PK-PD model simulations.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/farmacocinética , Inmunoglobulina G/farmacología , Modelos Biológicos , Investigación Biomédica Traslacional , Animales , Área Bajo la Curva , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Relación Dosis-Respuesta Inmunológica , Femenino , Humanos , Macaca fascicularis , Ratones , Ratones SCID , Resultado del Tratamiento , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Pharmacol Exp Ther ; 363(1): 35-44, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28768817

RESUMEN

WHIM syndrome is a rare congenital immunodeficiency disease, named after its main clinical manifestations: warts, hypogammaglobulinemia, infections, and myelokathexis, which refers to abnormal accumulation of mature neutrophils in the bone marrow. The disease is primarily caused by C-terminal truncation mutations of the chemokine receptor CXCR4, giving these CXCR4-WHIM mutants a gain of function in response to their ligand CXCL12. Considering the broad functions of CXCR4 in maintaining leukocyte homeostasis, patients are panleukopenic and display altered immune responses, likely as a consequence of impairment in the differentiation and trafficking of leukocytes. Treatment of WHIM patients currently consists of symptom relief, leading to unsatisfactory clinical responses. As an alternative and potentially more effective approach, we tested the potency and efficacy of CXCR4-specific nanobodies on inhibiting CXCR4-WHIM mutants. Nanobodies are therapeutic proteins based on the smallest functional fragments of heavy chain antibodies. They combine the advantages of small-molecule drugs and antibody-based therapeutics due to their relative small size, high stability, and high affinity. We compared the potential of monovalent and bivalent CXCR4-specific nanobodies to inhibit CXCL12-induced CXCR4-WHIM-mediated signaling with the small-molecule clinical candidate AMD3100. The CXCR4-targeting nanobodies displace CXCL12 binding and bind CXCR4-wild type and CXCR4-WHIM (R334X/S338X) mutants and with (sub-) nanomolar affinities. The nanobodies' epitope was mapped to extracellular loop 2 of CXCR4, overlapping with the binding site of CXCL12. Monovalent, and in particular bivalent, nanobodies were more potent than AMD3100 in reducing CXCL12-mediated G protein activation. In addition, CXCR4-WHIM-dependent calcium flux and wound healing of human papillomavirus-immortalized cell lines in response to CXCL12 was effectively inhibited by the nanobodies. Based on these in vitro results, we conclude that CXCR4 nanobodies hold significant potential as alternative therapeutics for CXCR4-associated diseases such as WHIM syndrome.


Asunto(s)
Especificidad de Anticuerpos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/terapia , Receptores CXCR4/inmunología , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/uso terapéutico , Verrugas/inmunología , Verrugas/terapia , Células HEK293 , Humanos , Síndromes de Inmunodeficiencia/genética , Mutación , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Verrugas/genética
4.
Drug Discov Today Technol ; 20: 59-69, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27986226

RESUMEN

The blood-brain barrier (BBB) represents a major obstacle for the delivery and development of drugs curing brain pathologies. However, this biological barrier presents numerous endogenous specialized transport systems that can be exploited by engineered nanoparticles to enable drug delivery to the brain. In particular, conjugation of glutathione (GSH) onto PEGylated liposomes (G-Technology®) showed to safely enhance delivery of encapsulated drugs to the brain. Yet, understanding of the mechanism of action remains limited and full mechanistic understanding will aid in the further optimization of the technology. In order to elucidate the mechanism of brain targeting by GSH-PEG liposomes, we here demonstrate that the in vivo delivery of liposomal ribavirin is increased in brain extracellular fluid according to the extent of GSH conjugation onto the liposomes. In vitro, using the hCMEC/D3 human cerebral microvascular endothelial (CMEC) cell line, as well as primary bovine and porcine CMEC (and in contrast to non-brain derived endothelial and epithelial cells), we show that liposomal uptake occurs through the process of endocytosis and that the brain-specific uptake is also glutathione conjugation-dependent. Interestingly, the uptake mechanism is an active process that is temperature-, time- and dose-dependent. Finally, early endocytosis events rely on cytoskeleton remodeling, as well as dynamin- and clathrin-dependent endocytosis pathways. Overall, our data demonstrate that the glutathione-dependent uptake mechanism of the G-Technology involves a specific endocytosis pathway indicative of a receptor-mediated mechanism, and supports the benefit of this drug delivery technology for the treatment of devastating brain diseases.


Asunto(s)
Antivirales/administración & dosificación , Encéfalo/metabolismo , Glutatión/administración & dosificación , Polietilenglicoles/administración & dosificación , Ribavirina/administración & dosificación , Animales , Antivirales/farmacocinética , Transporte Biológico , Bovinos , Línea Celular , Células Cultivadas , Células Endoteliales/metabolismo , Glutatión/química , Glutatión/farmacocinética , Células HEK293 , Humanos , Liposomas , Masculino , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Ratas Wistar , Ribavirina/farmacocinética , Porcinos
5.
J Biol Chem ; 288(41): 29562-72, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23979133

RESUMEN

The chemokine receptor CXCR7, belonging to the membrane-bound G protein-coupled receptor superfamily, is expressed in several tumor types. Inhibition of CXCR7 with either small molecules or small interference (si)RNA has shown promising therapeutic benefits in several tumor models. With the increased interest and effectiveness of biologicals inhibiting membrane-bound receptors we made use of the "Nanobody platform" to target CXCR7. Previously we showed that Nanobodies, i.e. immunoglobulin single variable domains derived from naturally occurring heavy chain-only camelids antibodies, represent new biological tools to efficiently tackle difficult drug targets such as G protein-coupled receptors. In this study we developed and characterized highly selective and potent Nanobodies against CXCR7. Interestingly, the CXCR7-targeting Nanobodies displayed antagonistic properties in contrast with previously reported CXCR7-targeting agents. Several high affinity CXCR7-specific Nanobodies potently inhibited CXCL12-induced ß-arrestin2 recruitment in vitro. A wide variety of tumor biopsies was profiled, showing for the first time high expression of CXCR7 in head and neck cancer. Using a patient-derived CXCR7-expressing head and neck cancer xenograft model in nude mice, tumor growth was inhibited by CXCR7-targeting Nanobody therapy. Mechanistically, CXCR7-targeting Nanobodies did not inhibit cell cycle progression but instead reduced secretion of the angiogenic chemokine CXCL1 from head and neck cancer cells in vitro, thus acting here as inverse agonists, and subsequent angiogenesis in vivo. Hence, with this novel class of CXCR7 inhibitors, we further substantiate the therapeutic relevance of targeting CXCR7 in head and neck cancer.


Asunto(s)
Neoplasias de Cabeza y Cuello/inmunología , Receptores CXCR/inmunología , Anticuerpos de Dominio Único/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Arrestinas/inmunología , Arrestinas/metabolismo , Unión Competitiva/inmunología , Camélidos del Nuevo Mundo/inmunología , Línea Celular Tumoral , Quimiocina CXCL12/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Neoplasias de Cabeza y Cuello/patología , Neoplasias de Cabeza y Cuello/prevención & control , Humanos , Ratones , Ratones Desnudos , Células 3T3 NIH , Ensayo de Unión Radioligante , Receptores CXCR/genética , Receptores CXCR/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Anticuerpos de Dominio Único/farmacología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , beta-Arrestinas
6.
Blood ; 119(9): 2024-32, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22238325

RESUMEN

The chemokine receptor CXCR4 is a critical regulator of cell migration and serves as a coreceptor for HIV-1. The chemokine stromal cell derived factor-1, also known as CXCL12, binds to CXCR4 and exerts its biologic functions partly through the small guanosine triphosphate hydrolase (GTPase) Rac1 (ras-related C3 botulinum toxin substrate 1). We show in different cell types, including CD34(+) hematopoietic stem and progenitor cells, that inhibition of Rac1 causes a reversible conformational change in CXCR4, but not in the related receptors CXCR7 or CCR5. Biochemical experiments showed that Rac1 associates with CXCR4. The conformational change of CXCR4 on Rac1 inhibition blocked receptor internalization and impaired CXCL12-induced Gα(i) protein activation. Importantly, we found that the conformation adopted by CXCR4 after Rac1 inhibition prevents HIV-1 infection of both the U87-CD4-CXCR4 cell line and of primary peripheral blood mononuclear cells. In conclusion, our data show that Rac1 activity is required to maintain CXCR4 in the responsive conformation that allows receptor signaling and facilitates HIV-1 infection; this implies that Rac1 positively regulates CXCR4 function and identifies the Rac1-CXCR4 axis as a new target for preventing HIV-1 infection.


Asunto(s)
Receptores CXCR4/química , Proteína de Unión al GTP rac1/metabolismo , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Células HL-60 , Humanos , Péptidos/química , Péptidos/farmacología , Conformación Proteica/efectos de los fármacos , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/química
7.
Proc Natl Acad Sci U S A ; 107(47): 20565-70, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059953

RESUMEN

The important family of G protein-coupled receptors has so far not been targeted very successfully with conventional monoclonal antibodies. Here we report the isolation and characterization of functional VHH-based immunoglobulin single variable domains (or nanobodies) against the chemokine receptor CXCR4. Two highly selective monovalent nanobodies, 238D2 and 238D4, were obtained using a time-efficient whole cell immunization, phage display, and counterselection method. The highly selective VHH-based immunoglobulin single variable domains competitively inhibited the CXCR4-mediated signaling and antagonized the chemoattractant effect of the CXCR4 ligand CXCL12. Epitope mapping showed that the two nanobodies bind to distinct but partially overlapping sites in the extracellular loops. Short peptide linkage of 238D2 with 238D4 resulted in significantly increased affinity for CXCR4 and picomolar activity in antichemotactic assays. Interestingly, the monovalent nanobodies behaved as neutral antagonists, whereas the biparatopic nanobodies acted as inverse agonists at the constitutively active CXCR4-N3.35A. The CXCR4 nanobodies displayed strong antiretroviral activity against T cell-tropic and dual-tropic HIV-1 strains. Moreover, the biparatopic nanobody effectively mobilized CD34-positive stem cells in cynomolgus monkeys. Thus, the nanobody platform may be highly effective at generating extremely potent and selective G protein-coupled receptor modulators.


Asunto(s)
Anticuerpos/farmacología , Quimiotaxis/efectos de los fármacos , VIH-1 , Receptores CXCR4/inmunología , Replicación Viral/efectos de los fármacos , Animales , Anticuerpos/aislamiento & purificación , Antígenos CD34 , Bencilaminas , Sitios de Unión/genética , Células COS , Chlorocebus aethiops , Ciclamas , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Células HEK293 , Movilización de Célula Madre Hematopoyética , Compuestos Heterocíclicos , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Nat Cancer ; 3(4): 418-436, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35469014

RESUMEN

Patient-derived organoids (PDOs) recapitulate tumor architecture, contain cancer stem cells and have predictive value supporting personalized medicine. Here we describe a large-scale functional screen of dual-targeting bispecific antibodies (bAbs) on a heterogeneous colorectal cancer PDO biobank and paired healthy colonic mucosa samples. More than 500 therapeutic bAbs generated against Wingless-related integration site (WNT) and receptor tyrosine kinase (RTK) targets were functionally evaluated by high-content imaging to capture the complexity of PDO responses. Our drug discovery strategy resulted in the generation of MCLA-158, a bAb that specifically triggers epidermal growth factor receptor degradation in leucine-rich repeat-containing G-protein-coupled receptor 5-positive (LGR5+) cancer stem cells but shows minimal toxicity toward healthy LGR5+ colon stem cells. MCLA-158 exhibits therapeutic properties such as growth inhibition of KRAS-mutant colorectal cancers, blockade of metastasis initiation and suppression of tumor outgrowth in preclinical models for several epithelial cancer types.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias Glandulares y Epiteliales , Anticuerpos Biespecíficos/farmacología , Receptores ErbB/metabolismo , Humanos , Imidazoles , Neoplasias Glandulares y Epiteliales/metabolismo , Células Madre Neoplásicas/metabolismo , Organoides , Pirazinas , Receptores Acoplados a Proteínas G/metabolismo
9.
Mol Pharmacol ; 76(4): 692-701, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19570946

RESUMEN

Human herpesviruses (HHVs) are widespread pathogens involved in proliferative diseases, inflammatory conditions, and cardiovascular diseases. During evolution, homologs of human chemokine receptors were integrated into the HHV genomes. In addition to binding endogenous chemokines, these viral G protein-coupled receptors (vGPCRs) have acquired the ability to signal in a constitutive manner. Ligand-induced and ligand-independent and autocrine and paracrine signaling properties of vGPCRs modify the functions of the expressing cells and lead to transformation and escape from immune surveillance. Furthermore, cross-talk or heterodimerization with endogenous chemokine receptors represent other ways for vGPCRs to modify intracellular signaling and cellular functions. As such, these viral receptors seem to play a prominent role during viral pathogenesis and life cycle and thus represent innovative antiviral therapies.


Asunto(s)
Herpesviridae/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Aterosclerosis/metabolismo , Dimerización , Genoma Viral , Herpesviridae/genética , Humanos , Sistemas de Lectura Abierta , Receptor Cross-Talk , Receptores Acoplados a Proteínas G/genética , Transducción de Señal
10.
Gastroenterology ; 135(2): 529-38, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18522803

RESUMEN

BACKGROUND & AIMS: Several lines of evidence support a role for Toll-like receptor (TLR) signaling to protect the intestine from pathogenic infection. We hypothesized that TLR signaling at the level of the intestinal epithelium is critical for mucosal immune responses. METHODS: We generated transgenic mice that express a constitutively active form of TLR4 in the intestinal epithelium (V-TLR4 mice). Lamina propria cellularity was evaluated by immunostaining and flow cytometry. Immunoglobulin (Ig) A levels in the stool and serum were measured by enzyme-linked immunosorbent assay. Chemokine and cytokine expression were analyzed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: V-TLR4 transgenic mice reproduced normally and had a normal life span. Constitutive activity of TLR4 in the intestinal epithelium promoted recruitment of B cells and an increase in fecal IgA levels. Intestinal epithelial cells of V-TLR4 mice expressed higher levels of CCL20 and CCL28, chemokines known to be involved in B-cell recruitment, and of a proliferation-inducing ligand (APRIL), a cytokine that promotes T-cell-independent class switching of B cells to IgA. The changes in B-cell numbers and IgA levels were blocked by simultaneous expression in intestinal epithelial cells of M3, a herpes virus protein that binds and inhibits multiple chemokines. CONCLUSIONS: TLR signaling in the intestinal epithelial cells significantly elevated the production of IgA in the intestine. This effect was mediated by TLR-induced expression of a specific set of chemokines and cytokines that promoted both recruitment of B cells into the lamina propria and IgA class switching of B cells.


Asunto(s)
Linfocitos B/inmunología , Movimiento Celular , Inmunidad Mucosa , Inmunoglobulina A/metabolismo , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Antígenos CD4/genética , Antígenos CD4/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Heces/química , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/metabolismo , Receptor Toll-Like 4/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
11.
Cancer Cell ; 33(5): 922-936.e10, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29763625

RESUMEN

HER2-driven cancers require phosphatidylinositide-3 kinase (PI3K)/Akt signaling through HER3 to promote tumor growth and survival. The therapeutic benefit of HER2-targeting agents, which depend on PI3K/Akt inhibition, can be overcome by hyperactivation of the heregulin (HRG)/HER3 pathway. Here we describe an unbiased phenotypic combinatorial screening approach to identify a bispecific immunoglobulin G1 (IgG1) antibody against HER2 and HER3. In tumor models resistant to HER2-targeting agents, the bispecific IgG1 potently inhibits the HRG/HER3 pathway and downstream PI3K/Akt signaling via a "dock & block" mechanism. This bispecific IgG1 is a potentially effective therapy for breast cancer and other tumors with hyperactivated HRG/HER3 signaling.


Asunto(s)
Anticuerpos Biespecíficos/administración & dosificación , Inmunoglobulina G/administración & dosificación , Neoplasias/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-3/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Anticuerpos Biespecíficos/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Inmunoglobulina G/farmacología , Células MCF-7 , Ratones , Modelos Moleculares , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/química , Receptor ErbB-3/química , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Drug Target ; 22(5): 460-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24524555

RESUMEN

Partly due to poor blood-brain barrier drug penetration the treatment options for many brain diseases are limited. To safely enhance drug delivery to the brain, glutathione PEGylated liposomes (G-Technology®) were developed. In this study, in rats, we compared the pharmacokinetics and organ distribution of GSH-PEG liposomes using an autoquenched fluorescent tracer after intraperitoneal administration and intravenous administration. Although the appearance of liposomes in the circulation was much slower after intraperitoneal administration, comparable maximum levels of long circulating liposomes were found between 4 and 24 h after injection. Furthermore, 24 h after injection a similar tissue distribution was found. To investigate the effect of GSH coating on brain delivery in vitro uptake studies in rat brain endothelial cells (RBE4) and an in vivo brain microdialysis study in rats were used. Significantly more fluorescent tracer was found in RBE4 cell homogenates incubated with GSH-PEG liposomes compared to non-targeted PEG liposomes (1.8-fold, p < 0.001). In the microdialysis study 4-fold higher (p < 0.001) brain levels of fluorescent tracer were found after intravenous injection of GSH-PEG liposomes compared with PEG control liposomes. The results support further investigation into the versatility of GSH-PEG liposomes for enhanced drug delivery to the brain within a tolerable therapeutic window.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Portadores de Fármacos/química , Glutatión/química , Polietilenglicoles/química , Animales , Barrera Hematoencefálica/metabolismo , Línea Celular , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Estabilidad de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fluoresceínas , Colorantes Fluorescentes , Glutatión/administración & dosificación , Glutatión/farmacocinética , Inyecciones Intravenosas , Inyecciones Espinales , Liposomas , Microdiálisis , Tamaño de la Partícula , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacocinética , Ratas , Ratas Wistar , Distribución Tisular
14.
PLoS One ; 9(1): e82331, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416140

RESUMEN

Brain cancer is a devastating disease affecting many people worldwide. Effective treatment with chemotherapeutics is limited due to the presence of the blood-brain barrier (BBB) that tightly regulates the diffusion of endogenous molecules but also xenobiotics. Glutathione pegylated liposomal doxorubicin (2B3-101) is being developed as a new treatment option for patients with brain cancer. It is based on already marketed pegylated liposomal doxorubicin (Doxil®/Caelyx®), with an additional glutathione coating that safely enhances drug delivery across the BBB. Uptake of 2B3-101 by human brain capillary endothelial cells in vitro was time-, concentration- and temperature-dependent, while pegylated liposomal doxorubicin mainly remained bound to the cells. In vivo, 2B3-101 and pegylated liposomal doxorubicin had a comparable plasma exposure in mice, yet brain retention 4 days after administration was higher for 2B3-101. 2B3-101 was overall well tolerated by athymic FVB mice with experimental human glioblastoma (luciferase transfected U87MG). In 2 independent experiments a strong inhibition of brain tumor growth was observed for 2B3-101 as measured by bioluminescence intensity. The effect of weekly administration of 5 mg/kg 2B3-101 was more pronounced compared to pegylated liposomal doxorubicin (p<0.05) and saline (p<0.01). Two out of 9 animals receiving 2B3-101 showed a complete tumor regression. Twice-weekly injections of 5 mg/kg 2B3-101 again had a significant effect in inhibiting brain tumor growth (p<0.001) compared to pegylated liposomal doxorubicin and saline, and a complete regression was observed in 1 animal treated with 2B3-101. In addition, twice-weekly dosing of 2B3-101 significantly increased the median survival time by 38.5% (p<0.001) and 16.1% (p<0.05) compared to saline and pegylated liposomal doxorubicin, respectively. Overall, these data demonstrate that glutathione pegylated liposomal doxorubicin enhances the effective delivery of doxorubicin to brain tumors and could become a promising new therapeutic option for the treatment of brain malignancies.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/patología , Doxorrubicina/análogos & derivados , Sistemas de Liberación de Medicamentos , Glutatión/análogos & derivados , Animales , Peso Corporal/efectos de los fármacos , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/ultraestructura , Capilares/patología , Proliferación Celular/efectos de los fármacos , Doxorrubicina/sangre , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glutatión/sangre , Glutatión/farmacocinética , Glutatión/farmacología , Glutatión/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Polietilenglicoles/uso terapéutico , Análisis de Supervivencia , Factores de Tiempo , Distribución Tisular/efectos de los fármacos , Resultado del Tratamiento
15.
PLoS One ; 7(11): e48935, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145028

RESUMEN

Chronic activation of Wnt/ß-catenin signaling is found in a variety of human malignancies including melanoma, colorectal and hepatocellular carcinomas. Interestingly, expression of the HCMV-encoded chemokine receptor US28 in intestinal epithelial cells promotes intestinal neoplasia in transgenic mice, which is associated with increased nuclear accumulation of ß-catenin. In this study we show that this viral receptor constitutively activates ß-catenin and enhances ß-catenin-dependent transcription. Our data illustrate that this viral receptor does not activate ß-catenin via the classical Wnt/Frizzled signaling pathway. Analysis of US28 mediated signaling indicates the involvement of the Rho-Rho kinase (ROCK) pathway in the activation of ß-catenin. Moreover, cells infected with HCMV show significant increases in ß-catenin stabilization and signaling, which is mediated to a large extent by expression of US28. The modulation of the ß-catenin signal transduction pathway by a viral chemokine receptor provides alternative regulation of this pathway, with potential relevance for the development of colon cancer and virus-associated diseases.


Asunto(s)
Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Citomegalovirus/genética , Citomegalovirus/metabolismo , Células HEK293 , Humanos , Ratones , Células 3T3 NIH , Receptores Virales/genética , Receptores Virales/metabolismo , Transducción de Señal , Transcripción Genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
16.
Eur J Med Chem ; 51: 184-92, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22424612

RESUMEN

The chemokine receptor CXCR7 is an atypical G protein-coupled receptor as it preferentially signals through the ß-arrestin pathway rather than through G proteins. CXCR7 is thought to be of importance in cancer and the development of CXCR7-targeting ligands is of huge importance to further elucidate the pharmacology and the therapeutic potential of CXCR7. In the present study, we synthesized 24 derivatives based on a compound scaffold patented by Chemocentryx and obtained CXCR7 ligands with pK(i) values ranging from 5.3 to 8.1. SAR studies were supported by computational 3D Fingerprint studies, revealing several important affinity descriptors. Two key compounds (29 and 30, VUF11207 and VUF11403) were found to be high-potency ligands that induce recruitment of ß-arrestin2 and subsequent internalization of CXCR7, making them important tool compounds in future CXCR7 research.


Asunto(s)
Amidas/química , Técnicas de Química Sintética , Modelos Moleculares , Receptores CXCR/agonistas , Estireno/química , Estireno/farmacología , Células HEK293 , Humanos , Conformación Molecular , Relación Estructura-Actividad Cuantitativa , Estireno/síntesis química
17.
Sci Signal ; 3(133): ra58, 2010 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-20682912

RESUMEN

US28 is a viral G protein (heterotrimeric guanosine triphosphate-binding protein)-coupled receptor encoded by the human cytomegalovirus (HCMV). In addition to binding and internalizing chemokines, US28 constitutively activates signaling pathways linked to cell proliferation. Here, we show increased concentrations of vascular endothelial growth factor and interleukin-6 (IL-6) in supernatants of US28-expressing NIH 3T3 cells. Increased IL-6 was associated with increased activation of the signal transducer and activator of transcription 3 (STAT3) through upstream activation of the Janus-activated kinase JAK1. We used conditioned growth medium, IL-6-neutralizing antibodies, an inhibitor of the IL-6 receptor, and short hairpin RNA targeting IL-6 to show that US28 activates the IL-6-JAK1-STAT3 signaling axis through activation of the transcription factor nuclear factor kappaB and the consequent production of IL-6. Treatment of cells with a specific inhibitor of STAT3 inhibited US28-dependent [(3)H]thymidine incorporation and foci formation, suggesting a key role for STAT3 in the US28-mediated proliferative phenotype. US28 also elicited STAT3 activation and IL-6 secretion in HCMV-infected cells. Analyses of tumor specimens from glioblastoma patients demonstrated colocalization of US28 and phosphorylated STAT3 in the vascular niche of these tumors. Moreover, increased phospho-STAT3 abundance correlated with poor patient outcome. We propose that US28 induces proliferation in HCMV-infected tumors by establishing a positive feedback loop through activation of the IL-6-STAT3 signaling axis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Interleucina-6/metabolismo , Neoplasias/etiología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Virales/farmacología , Animales , Retroalimentación Fisiológica , Humanos , Janus Quinasa 1/metabolismo , Ratones , Células 3T3 NIH , Neoplasias/virología , Receptores de Quimiocina , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
J Clin Invest ; 120(11): 3969-78, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20978345

RESUMEN

US28 is a constitutively active chemokine receptor encoded by CMV (also referred to as human herpesvirus 5), a highly prevalent human virus that infects a broad spectrum of cells, including intestinal epithelial cells (IECs). To study the role of US28 in vivo, we created transgenic mice (VS28 mice) in which US28 expression was targeted to IECs. Expression of US28 was detected in all IECs of the small and large intestine, including in cells expressing leucine rich repeat containing GPCR5 (Lgr5), a marker gene of intestinal epithelial stem cells. US28 expression in IECs inhibited glycogen synthase 3ß (GSK-3ß) function, promoted accumulation of ß-catenin protein, and increased expression of Wnt target genes involved in the control of the cell proliferation. VS28 mice showed a hyperplastic intestinal epithelium and, strikingly, developed adenomas and adenocarcinomas by 40 weeks of age. When exposed to an inflammation-driven tumor model (azoxymethane/dextran sodium sulfate), VS28 mice developed a significantly higher tumor burden than control littermates. Transgenic coexpression of the US28 ligand CCL2 (an inflammatory chemokine) increased IEC proliferation as well as tumor burden, suggesting that the oncogenic activity of US28 can be modulated by inflammatory factors. Together, these results indicate that expression of US28 promotes development of intestinal dysplasia and cancer in transgenic mice and suggest that CMV infection may facilitate development of intestinal neoplasia in humans.


Asunto(s)
Mucosa Intestinal/patología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Ratones Transgénicos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Animales , Línea Celular , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Humanos , Ratones , Ratones Endogámicos C57BL
19.
Methods Enzymol ; 460: 151-71, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19446724

RESUMEN

Human cytomegalovirus (HCMV) is a widely spread herpesvirus that can have serious consequences in immunocompromised hosts. Interestingly, HCMV genome encodes for four viral G protein-coupled receptors (vGPCRs), namely, US27, US28, UL33, and UL78. Thus far, US28 and UL33 have been shown to activate signaling pathways in a ligand-independent manner. US28 is the best characterized vGPCR and has been shown to be potentially involved in the development of HCMV-related diseases. As such, detailed investigation of these viral GPCR is of importance in order to understand molecular events occurring during viral pathogenesis and the potential identification of novel therapeutic targets. Herewith, we describe several approaches to study these HCMV-encoded vGPCRs. Using molecular biology, tags can be introduced in the vGPCRs, which may facilitate the study of their protein expression with various techniques, such as microscopy, Western blotting, enzyme-linked immunosorbent assay (ELISA), and flow cytometry. Furthermore, radioligand binding studies can be performed to screen for ligands for vGPCRs, but also to study kinetics of internalization. We also describe several signal transduction assays that can evaluate the signaling activity of these vGPCRs. In addition, we discuss different proliferation assays and an in vivo xenograft model that were used to identify the oncogenic potential of US28. The study of these vGPCRs in their viral context can be examined using recombinant HCMV strains generated by bacterial artificial chromosome mutagenesis. Finally, we show how these mutants can be used in several pharmacological and biochemical assays.


Asunto(s)
Citomegalovirus/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Proteínas Virales/fisiología , Animales , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Receptores de Quimiocina/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Trasplante Heterólogo , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
Cancer Res ; 69(7): 2861-9, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19318580

RESUMEN

The human cytomegalovirus (HCMV), potentially associated with the development of malignancies, encodes the constitutively active chemokine receptor US28. Previously, we have shown that US28 expression induces an oncogenic phenotype both in vitro and in vivo. Microarray analysis revealed differential expression of genes involved in oncogenic signaling in US28-expressing NIH-3T3 cells. In particular, the expression of cyclooxygenase-2 (COX-2), a key mediator of inflammatory diseases and major determinant in several forms of cancer, was highly up-regulated. US28 induced increases in COX-2 expression via activation of nuclear factor-kappaB, driving the production of vascular endothelial growth factor. Also, in HCMV-infected cells, US28 contributed to the viral induction of COX-2. Finally, the involvement of COX-2 in US28-mediated tumor formation was evaluated using the COX-2 selective inhibitor Celecoxib. Targeting COX-2 in vivo with Celecoxib led to a marked delay in the onset of tumor formation in nude mice injected with US28-transfected NIH-3T3 cells and a reduction of subsequent growth by repressing the US28-induced angiogenic activity. Hence, the development of HCMV-related proliferative diseases may partially be ascribed to the ability of US28 to activate COX-2.


Asunto(s)
Transformación Celular Viral/genética , Ciclooxigenasa 2/metabolismo , Citomegalovirus/genética , Receptores de Quimiocina/biosíntesis , Proteínas Virales/biosíntesis , Animales , Celecoxib , Línea Celular , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/farmacología , Citomegalovirus/metabolismo , Inducción Enzimática , Humanos , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Células 3T3 NIH , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Pirazoles/farmacología , Receptores de Quimiocina/genética , Sulfonamidas/farmacología , Transcripción Genética , Transfección , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA