Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 307
Filtrar
1.
J Exp Med ; 191(2): 213-24, 2000 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-10637267

RESUMEN

Cellular interleukin 10s (cIL-10s) of human and murine origin have extensive sequence and structural homology to the Epstein-Barr virus BCRF-I gene product, known as viral IL-10 (vIL-10). Although these cytokines share many immunosuppressive properties, vIL-10 lacks several of the immunostimulatory activities of cIL-10 on certain cell types. The molecular and cellular bases for this dichotomy are not currently defined. Here, we show that the single amino acid isoleucine at position 87 of cIL-10 is required for its immunostimulatory function. Substitution of isoleucine in cIL-10 with alanine, which corresponds to the vIL-10 residue, abrogates immunostimulatory activity for thymocytes, mast cells, and alloantigenic responses while preserving immunosuppressive activity for inhibition of interferon gamma production and prolongation of cardiac allograft survival. Conversely, substitution of alanine with isoleucine in vIL-10 converts it to a cIL-10-like molecule with immunostimulatory activity. This single conservative residue alteration significantly affects ligand affinity for receptor; however, affinity changes do not necessarily alter specific activities for biologic responses in a predictable fashion. These results suggest complex regulation of IL-10 receptor-ligand interactions and subsequent biological responses. These results demonstrate that vIL-10 may represent a captured and selectively mutated cIL-10 gene that benefits viral pathogenesis by leading to ineffective host immune responses. The ability to manipulate the activity of IL-10 in either a stimulatory or suppressive direction may be of practical value for regulating immune responses for disease therapy, and of theoretical value for determining what aspects of IL-10 activity are important for normal T cell responses.


Asunto(s)
Interleucina-10/inmunología , Isoleucina/inmunología , Proteínas de la Leche , Alanina/inmunología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células CHO , Células COS , División Celular , Células Cultivadas , Cricetinae , Proteínas de Unión al ADN/metabolismo , Humanos , Tolerancia Inmunológica , Interleucina-10/química , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Datos de Secuencia Molecular , Receptores de Interleucina/inmunología , Receptores de Interleucina/metabolismo , Receptores de Interleucina-10 , Factor de Transcripción STAT1 , Factor de Transcripción STAT3 , Factor de Transcripción STAT5 , Homología de Secuencia de Aminoácido , Transactivadores/metabolismo
2.
Science ; 235(4791): 895-8, 1987 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-3580039

RESUMEN

Heterogeneity in the expression of tumor-associated antigens, as defined by the binding of monoclonal antibodies, is a characteristic common to most, if not all, human carcinoma cell populations. Antigen-negative cells within the population can escape detection and therapy by their failure to bind the appropriate antibody. Therefore, the extent of antigenic heterogeneity is an important consideration when designing protocols for the management of cancer by administration of monoclonal antibodies. One approach to counteracting the effect of antigenic heterogeneity is the use of clone A of recombinant human leukocyte interferon (Hu-IFN-alpha A). Administration of Hu-IFN-alpha A in vivo effectively increased the amount of tumor antigen expressed by a human colon xenograft in situ and augmented the localization of a radiolabeled monoclonal antibody to the tumor site. Concomitant administration of Hu-IFN-alpha A and monoclonal antibody may thus be effective in overcoming the antigenic heterogeneity of carcinoma cell populations and in enhancing the efficacy of monoclonal antibodies in the detection and treatment of carcinoma lesions.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antígenos de Neoplasias/inmunología , Carcinoma/inmunología , Neoplasias del Colon/inmunología , Interferón Tipo I/administración & dosificación , Animales , Antígenos de Superficie/inmunología , Antígenos HLA/inmunología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias
3.
Science ; 202(4374): 1289-90, 1978 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-725605

RESUMEN

One of the species of human interferon produced by incubation of leukocytes with Newcastle disease virus was purified to homogeneity. It exhibited one peak of activity coinciding with a single protein band on sodium dodecyl sulfate-polyacrylamide gel electrophoresis.


Asunto(s)
Interferones/aislamiento & purificación , Leucocitos/análisis , Humanos , Métodos
4.
Curr Top Microbiol Immunol ; 316: 23-37, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17969442

RESUMEN

Interferon alpha (IFN-alpha) was approved by the US Food and Drug Administration on June 5, 1986 and paved the way for development of many other cytokines and growth factors. Nevertheless, we have barely touched the surface of understanding the multitude of human IFNs. This paper reviews the history of the purification of human leukocyte IFN, the cloning of the IFN-alphas, and the current state of knowledge of human interferon alpha genes and proteins.


Asunto(s)
Interferón-alfa/genética , Interferón-alfa/aislamiento & purificación , Cromatografía Líquida de Alta Presión , Clonación Molecular , ADN/química , ADN/genética , Historia del Siglo XX , Humanos , Interferón-alfa/química , Interferón-alfa/historia
5.
Mol Cell Biol ; 17(2): 695-706, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9001223

RESUMEN

Cell lines that are mutated in interferon (IFN) responses have been critical in establishing an essential role for the JAK family of nonreceptor tyrosine kinases in interferon signalling. Mutant gamma1A cells have previously been shown to be complemented by overexpression of JAK2. Here, it is shown that these cells carry a defect in, and can also be complemented by, the beta-subunit of the IFN-gamma receptor, consistent with the hypothesis that the mutation in these cells affects JAK2-receptor association. In contrast, mutant gamma2A cells lack detectable JAK2 mRNA and protein. By using gamma2A cells, the role of various domains and conserved tyrosine residues of JAK2 in IFN-gamma signalling was examined. Individual mutation of six conserved tyrosine residues, mutation of a potential phosphatase binding site, or mutation of the arginine residue in the proposed SH2-like domain had no apparent effect on signalling in response to IFN-gamma. Results with deletion mutants, however, indicated that association of JAK2 with the IFN-gammaR2 subunit requires the amino-terminal region but not the pseudokinase domain. Consistent with this, in chimeras with JAK1, the JAK2 amino-terminal region was required for receptor association and STAT1 activation. Conversely, a JAK1-JAK2 chimera with the amino-terminal domains of JAK1 linked to the pseudokinase and kinase domains of JAK2 is capable of reconstituting JAK-STAT signalling in response to IFN-alpha and -gamma in mutant U4C cells lacking JAK1. The specificity of the JAKs may therefore lie mainly in their structural interaction with different receptor and signalling proteins rather than in the substrate specificity of their kinase domains.


Asunto(s)
Interferón-alfa/farmacología , Interferón gamma/farmacología , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Antígenos CD/genética , Antígenos CD/metabolismo , Secuencia de Bases , Línea Celular , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Mutación del Sistema de Lectura , Humanos , Interleucina-6/farmacología , Janus Quinasa 1 , Janus Quinasa 2 , Datos de Secuencia Molecular , Mutación , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , ARN Mensajero/genética , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Proteínas Recombinantes de Fusión , Proteínas Recombinantes , Factor de Transcripción STAT1 , Transactivadores/metabolismo , Receptor de Interferón gamma
6.
Cytokine Growth Factor Rev ; 8(3): 189-206, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9462485

RESUMEN

With the purification and cloning of the interferon gamma (IFN-gamma) receptor chains the mechanism of IFN-gamma action and the resultant signal transduction events were delineated in remarkable detail. The interferon gamma (IFN-gamma) receptor complex consists of two chains: IFN-gammaR1, the ligand-binding chain, and IFN-gammaR2, the accessory chain. Binding of IFN-gamma causes oligomerization of the two IFN-gamma receptor subunits, IFN-gammaR1 and IFN-gammaR2, which initiates the signal transduction events: activation of Jak1 and Jak2 receptor associated protein tyrosine kinases, phosphorylation of the IFN-gammaR1 intracellular domain on Tyr440 followed by phosphorylation and activation of Stat1alpha, the latent transcriptional factor. With all these steps established, the IFN-gamma receptor complex has provided the basic model for understanding the receptors for other members of the family of class II cytokine receptors.


Asunto(s)
Interferón gamma/metabolismo , Receptores de Interferón/genética , Receptores de Interferón/fisiología , Animales , Humanos , Modelos Biológicos
7.
J Natl Cancer Inst ; 81(7): 502-12, 1989 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-2466127

RESUMEN

Freshly isolated cells from patients with pleural or peritoneal effusions cytologically diagnosed as adenocarcinoma (n = 43), malignant nonepithelial neoplasms (n = 10), and benign (n = 8) were analyzed for expression of constitutive levels of the tumor antigens TAG-72 [recognized by monoclonal antibody (MAb) B72.3] and carcinoembryonic antigen (CEA) (recognized by MAb COL-4) as well as the class I and class II major histocompatibility (MHC) antigens, and the ability of human interferons (Hu-IFNs) to enhance cell surface expression of those antigens as measured by MAb binding. Both type I and type II IFNs enhanced the expression of TAG-72 and CEA and altered the level of expression of the MHC antigens. Comparative studies of three different Hu-IFNs (IFN-alpha A, IFN-beta ser, and IFN-gamma) revealed that IFN-gamma was the most potent in augmenting either B72.3 or COL-4 binding. Unlike the IFN-gamma -mediated induction of the class II human leukocyte antigens, the change in tumor antigen expression consisted of enhanced constitutive antigen expression; de novo induction of either TAG-72 or CEA could not be achieved by either type I or type II IFN. Of 43 effusions isolated from different adenocarcinoma patients, 42 (97.7%) expressed either CEA or TAG-72, and treatment with Hu-IFN increased the level of expression of either antigen in 36 of 42 samples (85.7%). These studies demonstrate the augmentation of tumor-associated antigens on human carcinoma cells isolated from serous effusions by Hu-IFNs which may be used to enhance the targeting of conjugated MAbs to human carcinoma lesions.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos de Neoplasias/biosíntesis , Antígenos de Superficie/biosíntesis , Interferón beta , Interferones/farmacología , Anticuerpos Monoclonales , Antígeno Carcinoembrionario/biosíntesis , Exudados y Transudados/citología , Exudados y Transudados/inmunología , Glicoproteínas/biosíntesis , Humanos , Interferón Tipo I/farmacología , Interferón beta-1a , Interferon beta-1b , Interferón gamma/farmacología , Radioinmunoensayo , Proteínas Recombinantes/farmacología
8.
Cancer Res ; 56(18): 4250-4, 1996 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-8797600

RESUMEN

A phosphorylation site was introduced into chimeric monoclonal antibody B72.3 (MAb-chB72.3) by site-specific mutation of the coding sequence. The phosphorylation site for the cAMP-dependent protein kinase was positioned at the carboxyl terminus of the heavy-chain constant region of MAb-chB72.3. The resultant modified MAb-chB72.3-P was expressed in 293 cells and purified. The MAb-chB72.3-P protein was phosphorylated by the catalytic subunit of cAMP-dependent protein kinase with [gamma-32P]ATP to high radiospecific activity. The 32P-labeled MAb-chB72.3-P protein bound to cells expressing the tumor-associated glycoprotein 72 antigen. The introduction of phosphorylation sites into MAbs provides a new type of MAb for the diagnosis and treatment of cancers.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Anticuerpos Antineoplásicos/metabolismo , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glicoproteínas/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Antineoplásicos/biosíntesis , Secuencia de Bases , Línea Celular , Cartilla de ADN , Femenino , Duplicado del Terminal Largo de VIH , VIH-1/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/metabolismo , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/biosíntesis , Transfección
9.
Cancer Res ; 46(10): 4984-90, 1986 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2944575

RESUMEN

Human leukocyte (alpha) interferon (IFN-alpha) is composed of a multigene family within which at least eight different species have been expressed in Escherichia coli, isolated, and shown to exert a wide range of biological activities on different human target cells. In this study we utilized eight species of IFN-alpha (A, B, C, D, F, I, J, and K) and investigated their respective capabilities to alter the proliferation of a human breast carcinoma cell line (MCF-7). The antigens studied were all constitutively expressed on the MCF-7 cell surface: the Mr 180,000 carcinoembryonic antigen; a high molecular weight (greater than 10(6] glycoprotein, termed tumor-associated glycoprotein 72; and a major HLA histocompatibility antigen. The level of expression of each antigen was measured by the binding of monoclonal antibodies B1.1, B72.3, and W6/32, respectively. A high degree of diversity was found among the various IFN-alpha species with respect to their ability to enhance antigen expression and inhibit MCF-7 cell growth. The two most potent species, IFN-alpha A and IFN-alpha B, were found to increase the expression of tumor antigens as well as the HLA determinant by 2-5-fold. In contrast, IFN-alpha D and IFN-alpha J were virtually inactive in altering antigen expression but did inhibit the growth of MCF-7 cells. The remaining IFN-alpha species, -alpha C, -alpha F, -alpha I, and -alpha K, exerted an intermediate range of activities for both antigen enhancement and inhibition of MCF-7 cell growth. The relative ability of each species of IFN-alpha to inhibit MCF-7 cell growth appeared to be independent of their effectiveness in augmenting antigen expression. IFN-alpha D and IFN-alpha J, the two species that failed to alter tumor antigen expression, did, however, seem to interact with the interferon receptor since they inhibited MCF-7 cell growth and competed with other IFN-alpha species for the increase in carcinoembryonic antigen, tumor-associated glycoprotein 72, or HLA expression. A comparison of the concentrations of each IFN-alpha necessary to enhance antigen expression revealed that the surface HLA determinant was approximately 10-fold more sensitive to enhancement than was the tumor antigen, carcinoembryonic antigen. The individual members of the IFN-alpha family thus differ extensively in their ability to alter the level of antigen expression on the surface of MCF-7 breast carcinoma cells.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Antígenos de Neoplasias/análisis , Antígenos de Superficie/análisis , Antígeno Carcinoembrionario/análisis , Antígenos HLA/análisis , Interferón Tipo I/farmacología , Anticuerpos Monoclonales/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular , Femenino , Humanos , Receptores Inmunológicos/efectos de los fármacos , Receptores de Interferón , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad
10.
Cancer Res ; 48(20): 5674-9, 1988 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-3167826

RESUMEN

Using an assay based on the binding of a carcinoembryonic antigen (CEA)-specific monoclonal antibody, we have examined the expression of carcinoembryonic antigen genes in human colon tumor and normal fibroblast cell lines. CEA expression was not detectable in the normal fibroblast cell lines, whereas varying levels of high CEA expression were found in the colon tumor cell lines LS-174T, GEO, and WIDR. We have used a 550-base pair CEA probe derived from cloned complementary DNA to carry out Southern analysis of the DNA isolated from the normal and colon tumor cell lines. At high stringency, the CEA probe detected seven BamHI fragments in all DNAs analyzed. At low stringency, however, 14 BamHI fragments ranging from 1.5 to 23 kilobases were detected. Results of the Southern analysis demonstrate no amplification or rearrangement of the CEA genes in tumor cells. We used methylation-sensitive restriction endonucleases, HpaII and HhaI, to compare the degree of methylation of CEA family of genes in normal and colon tumor cell lines. Our results demonstrate that the CEA family of genes exists in a state of hypermethylation in the normal cell lines. In contrast, the CEA gene(s) are relatively hypomethylated in the tumor cell lines, suggesting a correlation between the state of methylation and degree of expression of the CEA gene(s). A comparison of the state of methylation of the CEA gene(s) in cells before and after treatment with the gamma-interferon (which up-regulates CEA steady-state mRNA levels) showed no detectable difference in the degree of DNA methylation. The segments of CEA genes that are hypermethylated in normal cells, but are hypomethylated in tumor cells, were also identified. Thus, these studies may help identify the sites of methylation that are crucial for the control of CEA gene regulation.


Asunto(s)
Antígeno Carcinoembrionario/genética , Carcinoma/genética , Neoplasias del Colon/genética , ADN/metabolismo , Anticuerpos Monoclonales , Antígeno Carcinoembrionario/biosíntesis , Carcinoma/metabolismo , Línea Celular , Neoplasias del Colon/metabolismo , Desoxirribonucleasa BamHI/metabolismo , Electroforesis en Gel de Poliacrilamida , Fibroblastos/metabolismo , Humanos , Metilación
11.
Cancer Res ; 49(10): 2651-5, 1989 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-2496918

RESUMEN

Recombinant human interferons have recently been shown to enhance tumor antigen expression, including carcinoembryonic antigen (CEA), on the surface of human carcinoma cells, which results in an increase in the targeting of antitumor monoclonal antibodies (MAb) in vivo. We report here the effect of recombinant human gamma-interferon (HuIFN-gamma) on the expression of human CEA and its related transcripts in several human colon carcinoma and normal human fibroblast cell lines. The colon tumor cell lines HT-29, WiDr, and LS-174T were each shown to express different constitutive levels of CEA glycopeptide, as measured by the binding of the CEA-specific MAb COL-4. Treatment with HuIFN-gamma enhanced the level of binding of COL-4 in total cell extracts of HT-29 and WiDr cells 2.5- and 6.5-fold, respectively. Using a CEA complementary DNA probe, this increase in MAb binding was shown to be accompanied by a 6- to 11-fold increase in the steady state levels of three CEA transcripts with sizes of 4.2, 3.5, and 2.8 kilobases. On the other hand, HuIFN-gamma treatment had no effect on the level of COL-4 binding or expression of CEA transcripts in LS-174T colon carcinoma cells, which are high constitutive expressors of CEA glycoprotein. Normal human fibroblast cell lines MRC-5 and WI38 had no detectable cytoplasmic CEA glycopeptide levels nor did they contain detectable levels of CEA mRNA, either before or after treatment with HuIFN-gamma. In contrast, HuIFN-gamma induced the de novo expression of the normal major histocompatibility complex class II antigen, HLA-DR, on HT-29 and WiDr colon cancer cells as well as the two fibroblast cell lines. Treatment of the LS-174T cell line with HuIFN-gamma did not result in the induction of class II HLA-DR antigen. These observations suggest that some common factors may be involved in the regulation of the CEA and class II histocompatibility genes. In addition, the demonstration that HuIFN-gamma enhances CEA expression in some carcinoma cell lines but fails to induce de novo expression of CEA transcripts in fibroblasts supports the potential application of HuIFN-gamma in enhancement of tumor targeting of antitumor MAbs and adds to our understanding of the mechanism of gamma-interferon-mediated up-regulation of some tumor antigens.


Asunto(s)
Antígeno Carcinoembrionario/genética , Carcinoma/análisis , Neoplasias del Colon/análisis , Interferón gamma/farmacología , ARN Mensajero/análisis , Antígeno Carcinoembrionario/análisis , Relación Dosis-Respuesta a Droga , Antígenos HLA-DR/análisis , Antígenos HLA-DR/genética , Humanos , Proteínas Recombinantes , Transcripción Genética , Células Tumorales Cultivadas
12.
Cancer Res ; 44(8): 3208-14, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6744259

RESUMEN

Treatment of human breast or colon carcinoma cells with recombinantly derived human leukocyte (clone A) interferon (IFN-alpha A) increases the surface expression of specific tumor-associated antigens (TAAs) recognized by monoclonal antibodies (MAbs). The MAbs used, B1.1, B6.2, and B72.3, recognize three distinct TAAs, i.e., the Mr 180,000 carcinoembryonic antigen, a Mr 90,000, and a Mr 220,000 to 400,000 glycoprotein, respectively. The binding of the MAbs to the surface of tumor cells increased in a dose-dependent manner, with optimal levels of TAA enhancement at 100 to 1,000 units IFN-alpha A/ml. Higher concentrations of IFN-alpha A that were cytostatic or cytotoxic were also less effective in enhancing TAA expression. Human melanoma (A375) cells and normal fibroblasts (WI-38 and Flow 4000) do not express any of the three TAAs, either before or after interferon treatment. The ability of IFN-alpha A to increase the expression of TAAs on human carcinoma cells was also temporally dependent, with optimal enhancement occurring after 16 to 24 hr. The enhancement of specific TAAs at the surface of the carcinoma cells by IFN-alpha A was confirmed, using fluorescence-activated cell sorter analysis. These data demonstrate that the IFN-alpha A-mediated increase of surface antigen is a result of both an accumulation of more antigen per cell, and an increase in the percentage of cells expressing the antigen. The ability of recombinant interferon to enhance specific TAAs on human carcinoma cells may be exploited in designing protocols for the in situ detection and therapy of human carcinoma lesions by MAbs, as well as in further defining the role of specific TAAs in the expression of the transformed phenotype.


Asunto(s)
Antígenos de Neoplasias/aislamiento & purificación , Antígenos de Superficie/aislamiento & purificación , Neoplasias de la Mama/inmunología , Neoplasias del Colon/inmunología , Interferón Tipo I/inmunología , Anticuerpos Monoclonales , Antígenos de Neoplasias/genética , Antígenos de Superficie/genética , Antígeno Carcinoembrionario/aislamiento & purificación , Femenino , Citometría de Flujo , Humanos , Interferón Tipo I/genética , Peso Molecular , Radioinmunoensayo
13.
Cancer Res ; 50(17): 5328-32, 1990 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-2201435

RESUMEN

Expression vectors for human alpha-interferon (Hu-IFN-alpha) J1, a site-specific mutant [Ser116]Hu-IFN-alpha J1, and Hu-IFN-alpha J/C or Hu-IFN-alpha C/J hybrids were constructed and expressed in Escherichia coli. These interferons and others were purified by immunoaffinity chromatography with a monoclonal antibody against human alpha-interferon. Their antiviral activity and ability to stimulate natural killer cell activity were determined in comparison to several other human interferons. These results provide some insight into structure-activity relationships for stimulating natural killer cells and confirm our previous conclusions that antiviral activity cannot be used to predict other activities for an individual IFN-alpha species. The observations suggest that the tertiary structure rather than any specific linear sequence of amino acids regulates the ability of the interferons to stimulate natural killer cell activity.


Asunto(s)
Interferón Tipo I/genética , Interferón Tipo I/farmacología , Células Asesinas Naturales/inmunología , Mutación , Secuencia de Aminoácidos , Secuencia de Bases , Clonación Molecular , Citotoxicidad Inmunológica/efectos de los fármacos , Escherichia coli/genética , Humanos , Células Asesinas Naturales/efectos de los fármacos , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Plásmidos , Proteínas Recombinantes , Mapeo Restrictivo , Relación Estructura-Actividad
14.
Cancer Res ; 58(12): 2489-99, 1998 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-9635566

RESUMEN

IFNs were first described as potent antiviral agents 40 years ago, and recombinant IFN-alpha2a and IFN-alpha2b were approved for the treatment of hairy cell leukemia just 11 years ago. Today, alpha-IFNs are approved worldwide for the treatment of a variety of malignancies and virologic diseases. Although the exact mechanism of action of IFN-alpha in the treatment of such diseases is not fully understood, many advances have been made in the characterization of the physicochemical and diverse biological properties of this highly pleiotropic cytokine. Here we review recent developments in our understanding of the antiviral and immunoregulatory properties of IFN-alpha, the nature of the multisubunit IFN-alpha receptor, and the molecular mechanisms of signal transduction. Where available, we have included comparative data on recombinant alpha-IFNs derived from both naturally occurring and nonnaturally occurring synthetic genes. We also review clinical data and data on the side effects and antigenicity of different sources of recombinant alpha-IFNs in humans. These latter topics are of clinical interest, because they may potentially affect the efficacy of these various products. Hopefully, what is already known about IFN will prompt further exploration into the mechanism(s) of action of IFN-alpha and thus deliver new applications for this prototypic cytokine, whose full therapeutic potential is yet to be realized.


Asunto(s)
Antineoplásicos/farmacología , Antivirales/farmacología , Interferón Tipo I/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Antivirales/química , Antivirales/metabolismo , Antivirales/uso terapéutico , Unión Competitiva , Citocinas/antagonistas & inhibidores , Dinoprostona/fisiología , Predicción , Humanos , Interferón Tipo I/química , Interferón Tipo I/metabolismo , Interferón Tipo I/uso terapéutico , Neoplasias/tratamiento farmacológico , Conformación Proteica , Receptor de Interferón alfa y beta , Receptores de Interferón/efectos de los fármacos , Receptores de Interferón/metabolismo , Proteínas Recombinantes , Transducción de Señal , Resultado del Tratamiento , Virosis/tratamiento farmacológico
15.
Oncogene ; 19(21): 2557-65, 2000 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10851054

RESUMEN

Cells of the immune system communicate with each other to initiate, establish and maintain immune responses. The communication occurs through cell-to-cell contact or through a variety of intercellular mediators that include cytokines, chemokines, growth factors and hormones. In the case of cytokines, the signal is transmitted from the outside to the inside of a cell through cell surface receptors specific for each cytokine. At this step the signal is also decoded and amplified: ligand binding causes recruitment and/or activation of numerous cytoplasmic proteins. One cytokine can activate a number of signal transduction pathways leading to regulation of a wide array of biological activities. One of these pathways, the Jak-Stat pathway, is briefly reviewed here with respect to the class II cytokine receptors. Signal transduction through receptors for interferons Type I (IFN-alpha, IFN-beta, IFN-omega) and Type II (IFN-gamma), and interleukin 10 (IL-10) is described in detail. In addition, a complex between tissue factor (TF) and coagulation factor VIIa, and two new receptors related to the class II cytokine receptor family are discussed. Oncogene (2000).


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Janus Quinasa 1 , Datos de Secuencia Molecular , Receptores de Citocinas/química , Receptores de Interferón/química , Receptores de Interferón/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-10 , Factor de Transcripción STAT1 , Alineación de Secuencia
16.
Oncogene ; 20(48): 7051-63, 2001 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11704829

RESUMEN

Abnormalities in cellular differentiation are frequent occurrences in human cancers. Treatment of human melanoma cells with recombinant fibroblast interferon (IFN-beta) and the protein kinase C activator mezerein (MEZ) results in an irreversible loss in growth potential, suppression of tumorigenic properties and induction of terminal cell differentiation. Subtraction hybridization identified melanoma differentiation associated gene-7 (mda-7), as a gene induced during these physiological changes in human melanoma cells. Ectopic expression of mda-7 by means of a replication defective adenovirus results in growth suppression and induction of apoptosis in a broad spectrum of additional cancers, including melanoma, glioblastoma multiforme, osteosarcoma and carcinomas of the breast, cervix, colon, lung, nasopharynx and prostate. In contrast, no apparent harmful effects occur when mda-7 is expressed in normal epithelial or fibroblast cells. Human clones of mda-7 were isolated and its organization resolved in terms of intron/exon structure and chromosomal localization. Hu-mda-7 encompasses seven exons and six introns and encodes a protein with a predicted size of 23.8 kDa, consisting of 206 amino acids. Hu-mda-7 mRNA is stably expressed in the thymus, spleen and peripheral blood leukocytes. De novo mda-7 mRNA expression is also detected in human melanocytes and expression is inducible in cells of melanocyte/melanoma lineage and in certain normal and cancer cell types following treatment with a combination of IFN-beta plus MEZ. Mda-7 expression is also induced during megakaryocyte differentiation induced in human hematopoietic cells by treatment with TPA (12-O-tetradecanoyl phorbol-13-acetate). In contrast, de novo expression of mda-7 is not detected nor is it inducible by IFN-beta+MEZ in a spectrum of additional normal and cancer cells. No correlation was observed between induction of mda-7 mRNA expression and growth suppression following treatment with IFN-beta+MEZ and induction of endogenous mda-7 mRNA by combination treatment did not result in significant intracellular MDA-7 protein. Radiation hybrid mapping assigned the mda-7 gene to human chromosome 1q, at 1q 32.2 to 1q41, an area containing a cluster of genes associated with the IL-10 family of cytokines. Mda-7 represents a differentiation, growth and apoptosis associated gene with potential utility for the gene-based therapy of diverse human cancers.


Asunto(s)
Antígenos de Neoplasias/genética , Apoptosis/genética , Cromosomas Humanos Par 1/genética , Diterpenos , Genes , Sustancias de Crecimiento/genética , Interleucinas , Proteínas de Neoplasias/genética , Neoplasias/genética , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/aislamiento & purificación , Secuencia de Bases , Carcinoma/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , División Celular/genética , Clonación Molecular , Dimetilsulfóxido/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor , Glioblastoma/patología , Sustancias de Crecimiento/biosíntesis , Sustancias de Crecimiento/aislamiento & purificación , Células HL-60/metabolismo , Células HL-60/patología , Humanos , Interferón Tipo I/farmacología , Células K562/metabolismo , Células K562/patología , Masculino , Melanocitos/metabolismo , Melanoma/química , Melanoma/genética , Melanoma/patología , Datos de Secuencia Molecular , Peso Molecular , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/aislamiento & purificación , Especificidad de Órganos , Osteosarcoma/patología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas Recombinantes , Terpenos/farmacología , Acetato de Tetradecanoilforbol/farmacología , Transfección , Células Tumorales Cultivadas/patología
17.
J Leukoc Biol ; 70(6): 950-60, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11739558

RESUMEN

The heterodimeric interferon (IFN)-gamma receptor (IFN-gammaR) is formed of two chains. Here we show that the binding chain (IFN-gammaR1) was highly expressed on the membranes of T, B, and myeloid cells. Conversely, the transducing chain (IFN-gammaR2) was highly expressed on the surfaces of myeloid cells, moderately expressed on B cells, and poorly expressed on the surfaces of T cells. Differential cell membrane expression of IFN-gammaR2 determined the number of receptor complexes that transduced the IFN-gamma signal and resulted in a different response to IFN-gamma. After IFN-gamma stimulation, high IFN-gammaR2 membrane expression induced rapid activation of signal transducer and activator of transcription-1 (STAT-1) and high levels of interferon regulatory factor-1 (IRF-1), which then triggered the apoptotic program. By contrast, low cell membrane expression resulted in slow activation of STAT-1, lower levels of IRF-1, and induction of proliferation. Because the forced expression of IFN-gammaR2 on T cells switched their response to IFN-gamma from proliferative to apoptotic, we concluded that the surface expression of IFN-gammaR2 determines whether a cell stimulated by IFN-gamma undergoes proliferation or apoptosis.


Asunto(s)
Apoptosis/inmunología , Linfocitos B/inmunología , Células Mieloides/inmunología , Receptores de Interferón/inmunología , Linfocitos T/inmunología , Linfocitos B/citología , División Celular/inmunología , Células Cultivadas , Proteínas de Unión al ADN/inmunología , Humanos , Factor 1 Regulador del Interferón , Interferón gamma/inmunología , Células Mieloides/citología , Fosfoproteínas/inmunología , Factor de Transcripción STAT1 , Transducción de Señal/inmunología , Linfocitos T/citología , Transactivadores/inmunología , Receptor de Interferón gamma
18.
AIDS ; 7(5): 693-7, 1993 May.
Artículo en Inglés | MEDLINE | ID: mdl-8100421

RESUMEN

OBJECTIVE: To evaluate the efficacy of low-dose oral recombinant interferon-alpha (IFN-alpha A) on clinical parameters, body weight, CD4+ lymphocyte counts and natural killer cell cytolytic activity in HIV-infected patients. DESIGN: Blinded crossover trial with controls for the protein and diluent components of the drug preparation. SETTING: Medical school outpatient referral center. PATIENTS, PARTICIPANTS: Eight patients with HIV-1 infection and a CD4+ lymphocyte count between 150 and 600 x 10(6)/l. Concurrent use of zidovudine was permitted. INTERVENTIONS: Patients received (daily, by mouth) 10 ml of a study solution of 2.5% albumin for 6 weeks, 150 IU IFN-alpha A for 6 weeks, and normal saline for 6 weeks. MAIN OUTCOME MEASURES: After two baseline visits, clinical assessments, vital signs, body weight, and laboratory tests, including enumeration of number and percentage of CD4+ and CD8+ lymphocytes and natural killer cell cytolytic activity, were performed every 3 weeks. Complete physical examinations were conducted every 6 weeks. RESULTS: No significant clinical or laboratory changes were observed during treatment with IFN-alpha A. Peak CD4+ lymphocyte counts were achieved at baseline in one patient, during albumin treatment in two patients, during IFN-alpha A treatment in one patient, and during saline treatment in four patients. All patients remained HIV-seropositive. Treatments were well-tolerated. CONCLUSION: This blinded pilot study of orally administered IFN-alpha A (150 IU daily for 6 weeks) did not demonstrate clinical benefit in HIV-infected patients.


Asunto(s)
Infecciones por VIH/terapia , VIH-1 , Interferón-alfa/administración & dosificación , Administración Oral , Adulto , Linfocitos T CD4-Positivos , Citotoxicidad Inmunológica , Tolerancia a Medicamentos , Femenino , Infecciones por VIH/inmunología , Humanos , Interferón alfa-2 , Interferón-alfa/efectos adversos , Células Asesinas Naturales/inmunología , Recuento de Leucocitos , Masculino , Proteínas Recombinantes
19.
J Invest Dermatol ; 83(1 Suppl): 128s-136s, 1984 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-6376647

RESUMEN

The structural and functional complexity of the human interferon system has become increasingly evident. More than eight different alpha (leukocyte) interferons are expressed in induced human cells in culture. Many of these have been purified by a combination of methods, including high-performance liquid chromatography. Moreover, at least 12 different human leukocyte interferons have been cloned, and several have been efficiently expressed in Escherichia coli and other organisms. The availability of purified species of leukocyte interferon, both natural and recombinant, has allowed structural work to be done, including amino acid sequence determinations, chemical modification studies, and the crystallization of one species. The purified material has also been used for the production of monoclonal antibodies with various specificities that are proving invaluable in rapid assays and purification techniques. Testing of the purified species for their relative potency in antiviral, antiproliferative, and immunomodulatory assays has begun to demonstrate the functional uniqueness and diversity of the purified alpha interferons. Hybrid interferon genes have been synthesized by splicing together parts of various cloned interferon genes. The resulting hybrid proteins have been valuable in establishing structure/function relationships. In several cases, the functional properties of the hybrid protein were novel and unpredicted from the properties of the parental molecules.


Asunto(s)
Bacterias/genética , Interferón Tipo I/aislamiento & purificación , Secuencia de Aminoácidos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Clonación Molecular/métodos , Efecto Citopatogénico Viral , ADN/genética , ADN Bacteriano/genética , ADN Recombinante , Escherichia coli/genética , Regulación de la Expresión Génica , Humanos , Interferón Tipo I/genética , Interferón Tipo I/farmacología , Hibridación de Ácido Nucleico , Plásmidos , ARN Mensajero/genética
20.
Gene ; 267(2): 233-42, 2001 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-11313150

RESUMEN

Defects in growth control and differentiation occur frequently in human cancers. In the case of human melanoma cells, treatment with a combination of fibroblast interferon (IFN-beta) and the protein kinase C activator mezerein (MEZ) results in an irreversible loss of proliferative potential and tumorigenic properties with a concomitant induction of terminal differentiation. These changes in cellular properties are associated with an induction and suppression in specific subsets of genes that occur in a temporal manner. To identify the complete repertoire of gene changes occurring during melanoma reversion to a more differentiated state a number of molecular approaches are being used. These include, subtraction hybridization using temporally spaced cDNA libraries, random cDNA isolation and evaluation by reverse Northern blotting and high throughput microarray analysis of subtracted cDNA clones. In the present study we have used a novel approach, rapid subtraction hybridization (RaSH), to identify and clone an additional gene of potential relevance to cancer growth control and terminal cell differentiation. RaSH has identified a human ubiquitin-processing protease gene, HuUBP43, that is differentially expressed in melanoma cells as a function of treatment with IFN-beta or IFN-beta + MEZ. HuUBP43 is a type I interferon inducible gene that is upregulated in a diverse panel of normal and tumor cells when treated with IFN-beta via the JAK/STAT kinase pathway. This gene may contribute to the phenotypic changes induced by IFN-beta during growth arrest and differentiation in human melanoma cells and other cell types as well as the antiviral and growth inhibitory effects of interferon.


Asunto(s)
Clonación Molecular/métodos , Diterpenos , Endopeptidasas/genética , Melanoma/genética , Secuencia de Aminoácidos , Secuencia de Bases , Northern Blotting , Diferenciación Celular , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Interferón beta/farmacología , Masculino , Melanoma/patología , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico/métodos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/efectos de los fármacos , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Terpenos/farmacología , Distribución Tisular , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Ubiquitina Tiolesterasa
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda