Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Bioorg Med Chem Lett ; 28(10): 1964-1971, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29636220

RESUMEN

Germinal center kinase-like kinase (GLK, also known as MAP4K3) has been hypothesized to have an effect on key cellular activities, including inflammatory responses. GLK is required for activation of protein kinase C-θ (PKCθ) in T cells. Controlling the activity of T helper cell responses could be valuable for the treatment of autoimmune diseases. This approach circumvents previous unsuccessful approaches to target PKCθ directly. The use of structure based drug design, aided by the first crystal structure of GLK, led to the discovery of several inhibitors that demonstrate potent inhibition of GLK biochemically and in relevant cell lines.


Asunto(s)
Proteína Quinasa C-theta/metabolismo , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Sitios de Unión , Línea Celular , Humanos , Concentración 50 Inhibidora , Interleucina-2/metabolismo , Ratones , Ratones Noqueados , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Estructura Terciaria de Proteína , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacología , Relación Estructura-Actividad , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
2.
J Exp Med ; 204(8): 1911-22, 2007 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-17646407

RESUMEN

Unmethylated CpG-oligodeoxynucleotides (ODNs) are generally thought of as potent adjuvants with considerable therapeutic potential to enhance immune responses against microbes and tumors. Surprisingly, certain so-called stimulatory CpG-ODNs strongly inhibited the effector phase of inflammatory arthritis in the K/BxN serum transfer system, either preventively or therapeutically. Also unexpected was that the inhibitory influence did not depend on the adaptive immune system cells mobilized in an immunostimulatory context. Instead, they relied on cells of the innate immune system, specifically on cross talk between CD8 alpha(+) dendritic cells and natural killer cells, resulting in suppression of neutrophil recruitment to the joint, orchestrated through interleukin-12 and interferon-gamma. These findings highlight potential applications of CpG-ODNs and downstream molecules as antiinflammatory agents.


Asunto(s)
Artritis/terapia , Islas de CpG , Células Dendríticas/citología , Inmunoterapia/métodos , Inflamación/terapia , Células Asesinas Naturales/citología , Animales , Antiinflamatorios/farmacología , Células Dendríticas/inmunología , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Células Asesinas Naturales/inmunología , Ratones , Modelos Biológicos , Neutrófilos/metabolismo , Oligonucleótidos/química , Transducción de Señal
3.
J Infect Dis ; 204(1): 103-14, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21628664

RESUMEN

Progressive multifocal leukoencephalopathy (PML), a fatal demyelinating disease caused by JC virus (JCV) infection of oligodendrocytes, may develop in patients with immune disorders following reactivation of chronic benign infection. Mutations of JCV capsid viral protein 1 (VP1), the capsid protein involved in binding to sialic acid cell receptors, might favor PML onset. Cerebrospinal fluid sequences from 37/40 PML patients contained one of several JCV VP1 amino acid mutations, which were also present in paired plasma but not urine sequences despite the same viral genetic background. VP1-derived virus-like particles (VLPs) carrying these mutations lost hemagglutination ability, showed different ganglioside specificity, and abolished binding to different peripheral cell types compared with wild-type VLPs. However, mutants still bound brain-derived cells, and binding was not affected by sialic acid removal by neuraminidase. JCV VP1 substitutions are acquired intrapatient and might favor JCV brain invasion through abrogation of sialic acid binding with peripheral cells, while maintaining sialic acid-independent binding with brain cells.


Asunto(s)
Proteínas de la Cápside/genética , Virus JC/genética , Virus JC/patogenicidad , Leucoencefalopatía Multifocal Progresiva/patología , Mutación Missense , Receptores Virales/metabolismo , Tropismo Viral , Adulto , Líquido Cefalorraquídeo/virología , Femenino , Desarrollo Humano , Humanos , Virus JC/aislamiento & purificación , Masculino , Persona de Mediana Edad , Proteínas Nucleares , Proteína de la Leucemia Promielocítica , Factores de Transcripción , Proteínas Supresoras de Tumor , Acoplamiento Viral
4.
Front Cell Neurosci ; 14: 592005, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33473245

RESUMEN

Microglia are central nervous system (CNS) resident immune cells that have been implicated in neuroinflammatory pathogenesis of a variety of neurological conditions. Their manifold context-dependent contributions to neuroinflammation are only beginning to be elucidated, which can be attributed in part to the challenges of studying microglia in vivo and the lack of tractable in vitro systems to study microglia function. Organotypic brain slice cultures offer a tissue-relevant context that enables the study of CNS resident cells and the analysis of brain slice microglial phenotypes has provided important insights, in particular into neuroprotective functions. Here we use RNA sequencing, direct digital quantification of gene expression with nCounter® technology and targeted analysis of individual microglial signature genes, to characterize brain slice microglia relative to acutely-isolated counterparts and 2-dimensional (2D) primary microglia cultures, a widely used in vitro surrogate. Analysis using single cell and population-based methods found brain slice microglia exhibited better preservation of canonical microglia markers and overall gene expression with stronger fidelity to acutely-isolated adult microglia, relative to in vitro cells. We characterized the dynamic phenotypic changes of brain slice microglia over time, after plating in culture. Mechanical damage associated with slice preparation prompted an initial period of inflammation, which resolved over time. Based on flow cytometry and gene expression profiling we identified the 2-week timepoint as optimal for investigation of microglia responses to exogenously-applied stimuli as exemplified by treatment-induced neuroinflammatory changes observed in microglia following LPS, TNF and GM-CSF addition to the culture medium. Altogether these findings indicate that brain slice cultures provide an experimental system superior to in vitro culture of microglia as a surrogate to investigate microglia functions, and the impact of soluble factors and cellular context on their physiology.

5.
Antimicrob Agents Chemother ; 53(5): 1840-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19258267

RESUMEN

Progressive multifocal leukoencephalopathy (PML) is a rare but frequently fatal disease caused by the uncontrolled replication of JC virus (JCV), a polyomavirus, in the brains of some immunocompromised individuals. Currently, no effective antiviral treatment for this disease has been identified. As a first step in the identification of such therapy, we screened the Spectrum collection of 2,000 approved drugs and biologically active molecules for their anti-JCV activities in an in vitro infection assay. We identified a number of different drugs and compounds that had significant anti-JCV activities at micromolar concentrations and lacked cellular toxicity. Of the compounds with anti-JCV activities, only mefloquine, an antimalarial agent, has been reported to show sufficiently high penetration into the central nervous system such that it would be predicted to achieve efficacious concentrations in the brain. Additional in vitro experiments demonstrated that mefloquine inhibits the viral infection rates of three different JCV isolates, JCV(Mad1), JCV(Mad4), and JCV(M1/SVEDelta), and does so in three different cell types, transformed human glial (SVG-A) cells, primary human fetal glial cells, and primary human astrocytes. Using quantitative PCR to quantify the number of viral copies in cultured cells, we have also shown that mefloquine inhibits viral DNA replication. Finally, we demonstrated that mefloquine does not block viral cell entry; rather, it inhibits viral replication in cells after viral entry. Although no suitable animal model of PML or JCV infection is available for the testing of mefloquine in vivo, our in vitro results, combined with biodistribution data published in the literature, suggest that mefloquine could be an effective therapy for PML.


Asunto(s)
Antivirales/farmacología , Virus JC/efectos de los fármacos , Mefloquina/farmacología , Neuroglía/virología , Replicación Viral/efectos de los fármacos , Antivirales/química , Astrocitos/virología , Línea Celular Transformada , Células Cultivadas , Humanos , Virus JC/aislamiento & purificación , Virus JC/fisiología , Leucoencefalopatía Multifocal Progresiva/virología , Mefloquina/química , Pruebas de Sensibilidad Microbiana/métodos , Modelos Moleculares , Neuroglía/citología , Virus 40 de los Simios/patogenicidad
6.
J Leukoc Biol ; 82(1): 124-32, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17412915

RESUMEN

Excessive scarring or fibrosis is a common feature of a wide spectrum of diseases characterized by an exaggerated Th2 response. The TLR/IL-1 receptor (IL-1R)-related protein ST2 is expressed in a membrane-bound form selectively by Th2 cells and was shown to be indispensable for some in vivo Th2 responses. ST2 was also found to block TLR signaling. We addressed the impact of the ST2 pathway on fibrogenesis using a mouse model of hepatic injury and fibrosis induced by carbon tetrachloride (CCl(4)). We showed that cytokine production by intrahepatic lymphocytes from CCl(4)-injured liver is abrogated in the absence of TLR-4. Interfering with the ST2 pathway using an ST2-Fc fusion protein accelerated and enhanced hepatic fibrosis, paralleled by the increasing ex vivo secretion of Th2 cytokines IL-4, -5, -10, and -13 by intrahepatic lymphocytes of ST2-Fc-treated, CCl(4)-gavaged mice. Absence of IL-4/13 signaling in IL-4Ralpha-deficient mice obliterated this ST2-Fc effect on fibrogenesis. Moreover, depletion of CD4(+) T cells abrogated ST2-Fc-enhanced Th2 cytokines and accelerated fibrosis. Thus, ST2-Fc caused overproduction of Th2 cytokines by intrahepatic CD4(+) T cells, possibly by modifying TLR-4 signaling in injured liver. This ST2-Fc-driven Th2 response exacerbated CCl(4)-induced hepatic fibrosis.


Asunto(s)
Citocinas/biosíntesis , Fibrosis/etiología , Hepatopatías/inmunología , Proteínas de la Membrana/farmacología , Células Th2/inmunología , Receptor Toll-Like 4/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Tetracloruro de Carbono , Fibrosis/inducido químicamente , Fibrosis/inmunología , Fragmentos Fc de Inmunoglobulinas , Proteína 1 Similar al Receptor de Interleucina-1 , Hepatopatías/prevención & control , Ratones , Receptores de Interleucina , Proteínas Recombinantes de Fusión
7.
J Interferon Cytokine Res ; 37(1): 20-31, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27835061

RESUMEN

Because of its tumor-suppressive effect, interferon-based therapy has been used for the treatment of melanoma. However, limited data are available regarding the antitumor effects of pegylated interferons, either alone or in combination with approved anticancer drugs. We report that treatment of human WM-266-4 melanoma cells with peginterferon beta-1a induced apoptotic markers. Additionally, peginterferon beta-1a significantly inhibited the growth of human SK-MEL-1, A-375, and WM-266-4 melanoma xenografts established in immunocompromised mice. Peginterferon beta-1a regressed large, established WM-266-4 xenografts in nude mice. Treatment of SK-MEL-1 tumor-bearing mice with a combination of peginterferon beta-1a and the MEK inhibitor PD325901 ((R)-N-(2,3-dihydroxypropoxy)-3,4-difluoro-2-(2-fluoro-4-iodophenylamino)benzamide) significantly improved tumor growth inhibition compared with either agent alone. Examination of the antitumor activity of peginterferon beta-1a in combination with approved anticancer drugs in breast and renal carcinomas revealed improved antitumor activity in these preclinical xenograft models, as did the combination of peginterferon beta-1a and bevacizumab in a colon carcinoma xenograft model.


Asunto(s)
Antineoplásicos/farmacología , Interferón beta/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Polietilenglicoles/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos Inmunológicos/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Humanos , Interferón beta/administración & dosificación , Interferón beta/farmacocinética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Ratones Noqueados , Mutación , Neoplasias/genética , Polietilenglicoles/administración & dosificación , Polietilenglicoles/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
8.
MAbs ; 7(4): 681-92, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25879139

RESUMEN

Polyomavirus JC (JCV) is the causative agent of progressive multifocal leukoencephalopathy (PML), a rare and frequently fatal brain disease that afflicts a small fraction of the immune-compromised population, including those affected by AIDS and transplantation recipients on immunosuppressive drug therapy. Currently there is no specific therapy for PML. The major capsid viral protein 1 (VP1) involved in binding to sialic acid cell receptors is believed to be a key player in pathogenesis. PML-specific mutations in JCV VP1 sequences present at the binding pocket of sialic acid cell receptors, such as L55F and S269F, abolish sialic acid recognition and might favor PML onset. Early diagnosis of these PML-specific mutations may help identify patients at high risk of PML, thus reducing the risks associated with immunosuppressive therapy. As a first step in the development of such early diagnostic tools, we report identification and characterization of affinity reagents that specifically recognize PML-specific mutations in VP1 variants using phage display technology. We first identified 2 peptides targeting wild type VP1 with moderate specificity. Fine-tuning via selection of biased libraries designed based on 2 parental peptides yielded peptides with different, yet still moderate, bindinspecificities. In contrast, we had great success in identifying synthetic antibodies that recognize one of the PML-specific mutations (L55F) with high specificity from the phage-displayed libraries. These peptides and synthetic antibodies represent potential candidates for developing tailored immune-based assays for PML risk stratification in addition to complementing affinity reagents currently available for the study of PML and JCV.


Asunto(s)
Proteínas de la Cápside/inmunología , Virus JC/inmunología , Leucoencefalopatía Multifocal Progresiva/inmunología , Mutación , Péptidos/inmunología , Anticuerpos de Cadena Única/inmunología , Proteínas de la Cápside/genética , Humanos , Virus JC/genética , Leucoencefalopatía Multifocal Progresiva/genética , Péptidos/genética , Anticuerpos de Cadena Única/genética
9.
J Interferon Cytokine Res ; 22(8): 873-80, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12396726

RESUMEN

Interferon-beta (IFN-beta) induces various antiproliferative activities. In solid tumor cells, IFN-beta inhibits cell cycle progression, which mainly occurs as S phase accumulation. The IFN-beta-induced cell cycle effect has been implicated in the antitumor effect of combinations of IFN-beta and chemotherapeutic drugs. In this report, we characterized the viability of various human tumor cells in vitro after combination treatment with IFN-beta protein and the chemotherapeutic drugs, cis-platinum (II) diamine dichloride (cisplatin), 5-fluorouracil (5-FU), paclitaxel (Taxol) and gemcitabine. IFN-beta could significantly potentiate the cytotoxicity of these chemotherapeutic drugs. The potentiating effect was observed after pretreatment of tumor cells with IFN-beta but did not require the constant presence of IFN-beta. The potentiating effect correlated with the sensitivity of the tumor cells to the IFN-beta-induced cytotoxicity. Furthermore, chemotherapeutic drugs also potentiated the cytotoxicity of IFN-beta. We conclude that the cell cycle effect per se did not determine the ability of IFN-beta to potentiate the cytotoxicity of chemotherapeutic drugs. We suggest that the combination of local IFN-beta gene therapy with chemotherapy could be an effective cancer treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Interferón beta/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/patología , Carcinoma/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Cisplatino/farmacología , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Sinergismo Farmacológico , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacología , Humanos , Interferón beta/administración & dosificación , Neoplasias Pulmonares/patología , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Neoplasias del Cuello Uterino/patología , Gemcitabina
10.
J Interferon Cytokine Res ; 22(2): 173-88, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11911800

RESUMEN

We analyzed whether interferon-alpha 2b (IFN-alpha 2b) and IFN-beta 1a engage their common receptor to generate activated receptor complexes possessing distinct signaling properties. Human vascular endothelial cells (HUVEC) are 100-1000-fold more sensitive to IFN-beta 1a than to IFN-alpha 2b in in vitro assays. An nonarray-based expression profiling (GeneCalling) technology was employed to compare the patterns and levels of gene expression induced by these IFN as the broadest means by which signaling events could be measured. To distinguish subtype-related differences from dose-related effects, RNA was prepared from HUVEC treated with 50-5000 pg/ml of each IFN. The results showed that at 50 pg/ml IFN, only a subset of the genes induced by IFN-beta 1a were also induced by IFN-alpha 2b and that individual genes were induced to higher levels by IFN-beta 1a. In contrast, at 5000 pg/ml, both subtypes induced essentially identical sets of genes to similar levels of expression. No genes were seen to be induced uniquely by IFN-alpha 2b but not by IFN-beta 1a. The results show that the two IFN are intrinsically capable of inducing similar gene induction responses and do not provide evidence that they generate activated receptor complexes possessing distinct signaling properties. In contrast, the two IFN generate gene induction patterns that are both qualitatively and quantitatively distinct at subsaturating and potentially physiologically more relevant concentrations.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Interferón Tipo I/fisiología , Interferón-alfa/farmacología , Interferón beta/farmacología , Receptores de Interferón/fisiología , Venas Umbilicales , Antineoplásicos/farmacología , Células Cultivadas , Análisis por Conglomerados , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica/estadística & datos numéricos , Genes/efectos de los fármacos , Humanos , Interferón alfa-2 , Interferón beta-1a , Proteínas de la Membrana , Receptor de Interferón alfa y beta , Proteínas Recombinantes , Transducción de Señal/efectos de los fármacos
11.
J Interferon Cytokine Res ; 30(10): 777-85, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20836711

RESUMEN

Multiple sclerosis is a chronic autoimmune disease of the central nervous system for which a number of disease-modifying therapies are available, including interferon beta (Avonex®, Rebif®, and Betaseron/Betaferon®), glatiramer acetate (Copaxone®), and an anti-VLA4 monoclonal antibody (Tysabri®). Despite the availability and efficacy of these protein and peptide drugs, there remains a significant number of patients who are untreated, including those with relatively mild disease who choose not to initiate therapy, those wary of injections or potential adverse events associated with therapy, and those who have stopped therapy due to perceived lack of efficacy. Since these drugs have side effects that may affect a patient's decision to initiate and to remain on treatment, there is a need to provide a therapy that is safe and efficacious but that requires a reduced dosing frequency and hence a concomitant reduction in the frequency of side effects. Here we describe the development of a PEGylated form of interferon beta-1a that is currently being tested in a multicenter, randomized, double-blind, parallel-group, placebo-controlled study in relapsing multiple sclerosis patients, with the aim of determining the safety and efficacy of 125 microg administered via the subcutaneous route every 2 or 4 weeks.


Asunto(s)
Interferón beta/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Polietilenglicoles/química , Humanos , Interferón beta-1a , Interferón beta/efectos adversos , Interferón beta/inmunología , Esclerosis Múltiple/inmunología , Polietilenglicoles/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Recurrencia
12.
Bioconjug Chem ; 17(1): 179-88, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16417267

RESUMEN

PEGylation of IFN-alpha has been used successfully to improve the pharmacokinetic properties and efficacy of the drug. To prepare a PEGylated form of human interferon-beta-1a (IFN-beta-1a) suitable for testing in vivo, we have synthesized 20 kDa mPEG-O-2-methylpropionaldehyde and used it to modify the N-terminal alpha-amino group of the cytokine. The PEGylated protein retained approximately 50% of the activity of the unmodified protein and had significantly improved pharmacokinetic properties following intravenous administration in rats. The clearance and volume of distribution at steady state were reduced approximately 30-fold and approximately 4-fold, respectively, resulting in a significant increase in systemic exposure as determined by the area under the curve. The elimination half-life of the PEGylated protein was approximately 13-fold greater than for the unmodified protein. The unmodified and PEGylated proteins were tested for their ability to inhibit the formation of radially oriented blood vessels entering the periphery of human SK-MEL-1 melanoma tumors in athymic nude homozygous (nu/nu) mice. In a single dose comparison study, administration of 1 x 10(6) units of unmodified IFN-beta-1a resulted in a 29% reduction in vessel number, while 1 x 10(6) units of PEGylated IFN-beta-1a resulted in a 58% reduction. Both treatments resulted in statistically significant reductions in mean vessel number as compared to the vehicle (control)-treated mice, with the PEGylated IFN-beta-1a-treated mice showing a statistically significantly greater reduction in mean vessel number as compared to the unmodified IFN-beta-1a-treated mice. In a multiple versus single dose comparison study, daily administration of 1 x 10(6) units of unmodified IFN-beta-1a for 9 days resulted in a 51% reduction in vessel number, while a single dose of 1 x 10(6) units of the PEGylated protein resulted in a 66% reduction. Both treatments resulted in statistically significant reductions in mean vessel number as compared to the vehicle-treated mice, with the PEGylated IFN-beta-1a-treated mice showing a statistically significantly greater reduction in mean vessel number as compared to the unmodified IFN-beta-1a-treated mice. Therefore, the improved pharmacokinetic properties of the modified protein translated into improved efficacy. Since unmodified IFN-beta is used for the treatment of multiple sclerosis and hepatitis C virus infection, a PEGylated form of the protein such as 20 kDa mPEG-O-2-methylpropionaldehyde-modified IFN-beta-1a may serve as a useful adjunct for the treatment of these diseases. In addition, the antiangiogenic effects of PEGylated IFN-beta-1a may be harnessed for the treatment of certain cancers, either as a sole agent or in combination with other antitumor drugs.


Asunto(s)
Aldehídos/uso terapéutico , Antivirales/uso terapéutico , Interferón beta/uso terapéutico , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Polietilenglicoles/uso terapéutico , Aldehídos/síntesis química , Aldehídos/farmacocinética , Animales , Antivirales/química , Antivirales/farmacocinética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Virus de la Encefalomiocarditis/efectos de los fármacos , Femenino , Semivida , Humanos , Interferón beta-1a , Interferón beta/química , Interferón beta/farmacocinética , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Tasa de Depuración Metabólica , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Polietilenglicoles/síntesis química , Polietilenglicoles/farmacocinética , Ratas , Ratas Endogámicas Lew
13.
J Immunol ; 168(9): 4462-71, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11970990

RESUMEN

Alefacept, an immunomodulatory recombinant fusion protein composed of the first extracellular domain of LFA-3 fused to the human IgG1 hinge, C(H)2, and C(H)3 domains, has recently been shown in phase II and III clinical trials to safely reduce disease expression in patients with chronic plaque psoriasis. Alefacept modulates the function of and selectively induces apoptosis of CD2(+) human memory-effector T cells in vivo. We have sought to gain further understanding of the mechanisms of action that influence the biological activity of alefacept and may contribute to its efficacy and patient responsiveness. Specifically evaluated is the ability of alefacept to activate intracellular signals mediated via CD2 and/or Fc gamma RIII (CD16). Experimentation using isoforms of alefacept engineered to have amino acid substitutions in the IgG1 C(H)2 domain that impact Fc gamma R binding indicate that alefacept mediates cognate interactions between cells expressing human CD2 and CD16 to activate cells, e.g., increase extracellular signal-regulated kinase phosphorylation, up-regulate cell surface expression of the activation marker CD25, and induce release of granzyme B. In the systems used, this signaling is shown to require binding to CD2 and CD16 and be mediated through CD16, but not CD2. Experimentation using human CD2-transgenic mice and isoforms of alefacept confirmed the requirement for Fc gamma R binding for detection of the pharmacological effects of alefacept in vivo. Thus alefacept acts as an effector molecule, mediating cognate interactions to activate Fc gamma R(+) cells (e.g., NK cells) to induce apoptosis of sensitive CD2(+) target cells.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígenos CD2/metabolismo , Linfocitos/inmunología , Receptores de IgG/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Alefacept , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis , Antígenos CD2/análisis , Antígenos CD2/genética , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Perfilación de la Expresión Génica , Humanos , Interleucina-2/farmacología , Células Jurkat , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Receptores de IgG/genética , Receptores de IgG/inmunología , Transducción de Señal , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA