Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Colorectal Dis ; 12(6): 561-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19250260

RESUMEN

OBJECTIVE: Colorectal cancer is immunogenic. However, it is also associated with suppression of host immunity. Identifying the mechanisms involved in immune suppression is necessary to develop future immunotherapeutic strategies. The aim of this study was to assess immune cell function in colorectal cancer patients. METHOD: A total of 80 colorectal cancer patients (41 male) prior to treatment and 38 matched controls (21 male) were recruited. Venous blood samples were taken. White blood cell composition was determined using monoclonal antibodies. Levels of cytokines IFN-gamma, TNF-alpha, IL-2, IL-10, IL-4 and IL-6 were measured from the supernatants of activated peripheral blood mononuclear cells (PBMC) following thawing and re-suspension. Peripheral blood mononuclear proliferation was measured using 3H-Thymidine. RESULTS: Stage I-III cancer patients had elevated percentages of CD8 T cell (P = 0.004) whilst stage IV patients had low total lymphocyte percentages (P = 0.016). Monocyte and NKT cell percentage decreased with advanced tumour stages (P = 0.013 and P = 0.038). Patients had lower PBMC proliferation and production of the TH1 cytokines (IFN-gamma and TNF-alpha) (P < 0.001) than that of the controls. IL-6 and IL-4 production were not significantly different. IFN-gamma and TNF-alpha concentrations reduced with tumour vascular invasion (P = 0.011 and P = 0.019). CONCLUSION: Colorectal cancer induces an immunological response, shifting the cytokine balance. The most profound changes are seen once disease has spread systemically.


Asunto(s)
Neoplasias Colorrectales/inmunología , Citocinas/sangre , Células TH1/inmunología , Células Th2/inmunología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Inmunidad Celular , Huésped Inmunocomprometido , Inmunofenotipificación , Interferón gamma , Interleucinas/sangre , Masculino , Persona de Mediana Edad , Factor de Necrosis Tumoral alfa
6.
Curr Drug Targets CNS Neurol Disord ; 1(1): 17-30, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12769632

RESUMEN

Multiple sclerosis (MS) is a demyelinating disease of the central nervous system of unknown etiology. Two of the major therapies for the treatment of MS, interferon-beta and glatiramer acetate, show only limited evidence that long-term treatment slows disability. There is a great need for new drugs that will halt, reverse, and prevent the development of MS. This article reviews therapies currently in use and describes innovative strategies being developed to alter the disease course. New technologies in gene expression profiling offer hopeful directions toward the design of successful drug therapies and diagnostic testing for MS. Additionally, the new fields of genomics and proteomics offer the promise of novel treatments, and should help to reveal the mechanisms of disease initiation and pathological progression.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Terapia Genética/métodos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Animales , Sistemas de Liberación de Medicamentos/tendencias , Terapia Genética/tendencias , Genómica/métodos , Genómica/tendencias , Humanos , Esclerosis Múltiple/genética , Tecnología Farmacéutica/métodos , Tecnología Farmacéutica/tendencias
7.
Am J Pathol ; 159(4): 1219-24, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11583948

RESUMEN

Demyelination in multiple sclerosis and in animal models is associated with infiltrating CD8+ and CD4+ T cells. Although oligodendrocytes and axons are damaged in these diseases, the roles T cells play in the demyelination process are not completely understood. Antigen-specific CD8+ T cell lysis of target cells is dependent on interactions between the T cell receptor and major histocompatibility complex (MHC) class I-peptide complexes on the target cell. In the normal central nervous system, expression of MHC molecules is very low but often increases during inflammation. We set out to precisely define which central nervous system cells express MHC molecules in vivo during infection with a strain of murine hepatitis virus that causes a chronic, inflammatory demyelinating disease. Using double immunofluorescence labeling, we show that during acute infection with murine hepatitis virus, MHC class I is expressed in vivo by oligodendrocytes, neurons, microglia, and endothelia, and MHC class II is expressed only by microglia. These data indicate that oligodendrocytes and neurons have the potential to present antigen to T cells and thus be damaged by direct antigen-specific interactions with CD8+ T lymphocytes.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Virus de la Hepatitis Murina , Neuronas/metabolismo , Oligodendroglía/metabolismo , Enfermedades de la Médula Espinal/metabolismo , Animales , Infecciones por Coronavirus/patología , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Variación Genética , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/metabolismo , Virus de la Hepatitis Murina/genética , Enfermedades de la Médula Espinal/patología
8.
Lab Invest ; 79(2): 235-42, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10068211

RESUMEN

To recognize and respond immunologically to foreign antigens, T lymphocytes require the presentation of foreign peptides by MHC molecules. To determine which cells of the central nervous system (CNS) are capable of expressing MHC molecules, we used confocal microscopy and dual immunofluorescence with cell-specific and MHC-specific antibodies to study brain sections of adult mice. We took advantage of transgenic mice that initiate CNS-specific expression of IFN-gamma at 8 weeks of age. This inflammatory cytokine is a strong inducer of MHC expression both in culture and in vivo. From this analysis, we clearly found MHC class I and II expression on endothelial, microglial, and oligodendrocyte cell types, but did not find astrocytes or neurons capable of expressing either MHC class I or II molecules under these conditions. This finding suggests that, although microglia and oligodendrocytes may participate in the antigen presentation process in the organism, we found no in vivo evidence to support the concept that astrocytes act as antigen-presenting cells.


Asunto(s)
Sistema Nervioso Central/efectos de los fármacos , Interferón gamma/farmacología , Complejo Mayor de Histocompatibilidad/fisiología , Animales , Astrocitos/inmunología , Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Endotelio/citología , Endotelio/inmunología , Técnica del Anticuerpo Fluorescente , Antígenos de Histocompatibilidad Clase I/análisis , Antígenos de Histocompatibilidad Clase II/análisis , Interferón gamma/genética , Ratones , Ratones Transgénicos/genética , Microglía/inmunología , Microscopía Confocal , Neuronas/inmunología , Oligodendroglía/inmunología
9.
J Exp Med ; 184(6): 2371-84, 1996 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8976191

RESUMEN

One hypothesis for the etiology of central nervous system (CNS) autoimmune disease is that infection by a virus sharing antigenic epitopes with CNS antigens (molecular mimicry) elicits a virus-specific immune response that also recognizes self-epitopes. To address this hypothesis, transgenic mice were generated that express the nucleoprotein or glycoprotein of lymphocytic choriomeningitis virus (LCMV) as self in oligodendrocytes. Intraperitoneal infection with LCMV strain Armstrong led to infection of tissues in the periphery but not the CNS, and the virus was cleared within 7-14 d. After clearance, a chronic inflammation of the CNS resulted, accompanied by upregulation of CNS expression of MHC class I and II molecules. A second LCMV infection led to enhanced CNS pathology, characterized by loss of myelin and clinical motor dysfunction. Disease enhancement also occurred after a second infection with unrelated viruses that cross-activated LCMV-specific memory T cells. These findings indicate that chronic CNS autoimmune disease may be induced by infection with a virus sharing epitopes with a protein expressed in oligodendrocytes and this disease may be enhanced by a second infection with the same or an unrelated virus. These results may explain the association of several different viruses with some human autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/virología , Enfermedades del Sistema Nervioso Central/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Oligodendroglía/virología , Proteínas Virales/biosíntesis , Animales , Enfermedades Autoinmunes/inmunología , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Enfermedades del Sistema Nervioso Central/virología , Cartilla de ADN , Humanos , Inmunohistoquímica , Memoria Inmunológica , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodendroglía/patología , Reacción en Cadena de la Polimerasa , Médula Espinal/inmunología , Médula Espinal/patología , Médula Espinal/virología , Linfocitos T/inmunología , Linfocitos T/patología , Proteínas Virales/genética
10.
Immunol Rev ; 152: 111-43, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8930670

RESUMEN

Viruses have often been associated with autoimmune diseases. One mechanism by which self-destruction can be triggered is molecular mimicry. Many examples of cross-reactive immune responses between pathogens and self-antigens have been described. This review presents two transgenic models of autoimmune disease induced by a virus through activation of anti-self lymphocytes. Viral antigens are expressed as transgenes either in beta-cells of the pancreas or in the oligodendrocytes of the CNS. Infection by a virus encoding the same gene activated autoreactive T cells that cleared the viral infection, and as a consequence of transgene expression resulted in organ-specific autoimmune disease. In both transgenic mouse models, autoreactive lymphocytes that escaped thymic negative selection were present in the periphery. Several factors are described that play a role in the regulation of the self-reactive process precipitated by a viral infection. These include the quantity of activated autoreactive T cells, the affinity of these T cells, the number of memory T cells generated following primary infection, costimulation by accessory molecules, and the types and locations of cytokines produced. In addition, unique barriers exist in target tissues that prevent or suppress autoreactive responses and define to a large extent the outcome of disease. Restimulation of autoreactive memory lymphocytes may be required to bypass these barriers and enhance autoimmune disease. Therapy directed at modifying these factors can reduce and even prevent autoimmune disease after it has been initiated.


Asunto(s)
Enfermedades del Sistema Nervioso Central/inmunología , Sistema Nervioso Central/inmunología , Insulina/inmunología , Islotes Pancreáticos/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones Transgénicos , Proteína Básica de Mielina/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/virología , Enfermedades del Sistema Nervioso Central/virología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Modelos Animales de Enfermedad , Humanos , Insulina/genética , Ratones , Proteína Básica de Mielina/genética , Autotolerancia/inmunología , Virus/patogenicidad
11.
J Virol ; 70(3): 1799-803, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8627703

RESUMEN

Infidelity of genome applications of RNA viruses leads to the generation of viral quasispecies both in vitro and in vivo. However, the biological significance of such generated variants in vivo is largely unknown and controversial. To study this issue, we continued our evaluation of the tropism of a lymphocytic choriomeningitis virus (LCMV) variant termed clone 13 with its parental virus clonal pool ARM 53b (wild-type parent) for neuronal cells in vivo. Earlier in vivo and in vitro studies noted that the wild-type virus contained a Phe at glycoprotein (GP) residue 260 which correlated with neuron tropism compared with LCMV variants containing a Leu at residue 260 which showed selected tropism for cells of the immune system (C.F. Evans, P. Borrow, J. C. de la Torre, and M. B. A. Oldstone J. Virol. 68:7367-7373, 1994; L. Villarete, T. Somasundaram, and R. Ahmed, J. Virol 68:7490-7496, 1994). Here we (i) evaluated the ability of the viral variants with either a Phe or Leu at GP residue 260 to replicate in vivo in the spleen, liver, or brain, (ii) analyzed the ability of these viruses to compete against each other for cell (neuron)-specific selection following a single viral inoculation of different ratios of both viruses, and (iii) utilized genetic reassortants of both viruses to test their ability to replicate in neurons in vivo. We found that viral variants containing either a Phe or Leu at GP residue 260 were equally capable of replicating in neurons, but when inoculated together, neurons selected for the viral population containing Phe at GP residue 260 over viruses containing a Leu at this position. This was in contrast to selection in the liver and spleen that favored viruses with Leu and not Phe at GP residue 260. Analysis of inoculations with viral reassortants indicated that genes encoded on the short RNA (the GP and nucleoprotein, not the L [polymerase] and Z proteins that are encoded by the large RNA) were associated with neurotropism. Since the nucleoprotein sequences of wild-type Armstrong and clone 13 are identical, it is likely that specific cytoplasmic factors of the neurons play a fundamental role in the selection of virus with Phe at GP residue 260.


Asunto(s)
Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Neuronas/virología , Animales , Encéfalo/patología , Encéfalo/virología , Línea Celular , Cricetinae , Genes Virales , Glicoproteínas/genética , Glicoproteínas/metabolismo , Hígado/virología , Linfocitos , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Macrófagos , Ratones , Ratones Endogámicos BALB C , Virus ARN , ARN Viral , Especificidad de la Especie , Bazo/virología , Replicación Viral/genética
13.
Virology ; 211(2): 443-50, 1995 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-7645248

RESUMEN

The proteins of lymphocytic choriomeningitis virus (LCMV) contain only three known peptide regions that are processed and then held in place by the MHC class I H-2b (Db) glycoprotein on the cell's surface for recognition by LCMV-specific Db-restricted cytotoxic T lymphocytes (CTL). These peptides are from the glycoprotein (GP), amino acids 33-41 KAVYNFATC (GP1) and 276-286 SGVENPGGYCL (GP2), and the nucleoprotein (NP), 396-404. We have used CTL clones that recognized only GP1, GP2, and NP to select viral variants that upon infecting cells bearing H-2b molecules escaped recognition by virus-specific CTL directed against the viral GP (GP1 + GP2) mutant, termed GPV, or the viral GP and NP (GP1 + GP2 + NP) mutant, termed GPV+NPV. These CTL "escape" variants nevertheless elicited sufficient host-protective activity in vivo to abort acute infection and prevent the occurrence of persistent infection. This protection was CD8+ lymphocyte mediated and associated with the generation of a novel (for H-2b mice) CTL response to the viral L protein. Hence CTL epitopes form a hierarchy, in which responses to "weak" epitopes are suppressed in the presence of "stronger" epitopes. Mutation in the strong epitopes may be of limited biological significance since the host can mount a protective response directed against the second level (weak) epitopes.


Asunto(s)
Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase I/química , Antígenos de Histocompatibilidad Clase I/inmunología , Células Asesinas Naturales/inmunología , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Proteínas Virales/genética , Proteínas Virales/inmunología
14.
Virology ; 210(1): 29-40, 1995 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-7793078

RESUMEN

Cytotoxic T lymphocytes (CTL) play a pivotal role in preventing persistent viral infections and aborting acute infections. H-2Db-restricted CTL optimally recognize a specific peptide of 9 to 11 amino acids (aa) derived from a viral protein and held in place (restricted) by a MHC class I glycoprotein on the surfaces of infected cells. Only three peptide sequences with the appropriate Db motif from lymphocytic choriomeningitis virus Armstrong strain (LCMV) are known to be presented to CTL by H-2Db molecules; they are from the glycoproteins (GP), residues 33-41 KAVYNFATC (GP1) and 276-286 SGVENPGGYCL (GP2), and the nucleoprotein (NP), 396-404 FQPQNGQFI. Incubation of virally infected H-2b cells with CTL clones that recognize only GP1, GP2, or NP leads to the selection of viral variants which upon infecting cells bearing H-2b molecules, escape recognition by CTL of the appropriate specificity. Nucleic acid sequencing showed a single mutation in GP1 (aa 38 F-->L), GP2 (aa 282 G-->D), or NP (aa 403 F-->L) in the variant viruses. When wild-type (wt) LCMV peptides and the three variant peptides (GP1, GP2, NP) were synthesized and subjected to a competitive inhibition binding assay, no differences in binding affinity for H-2Db were found between the wt and variant peptides. Uninfected cells coated with the wt peptide were recognized and lysed by the appropriate CTL clone or by in vivo-primed bulk CTL, but similar targets coated with the GP1, GP2, or NP variant peptides were not. This result, coupled with computer graphic analysis of these variant peptides with the recently solved three-dimensional structure for the Db MHC class I molecule, placed the side chain of the mutated residues on the outer surface of the MHC-peptide complex and accessible to the T cell receptor. Ala substitution at GP residue 38 or 282 or at NP 403 also abrogated CTL recognition and lysis. Inoculation of any one of the mutated viral variants into mice produced an effective CTL response to the other two nonmutated GP or NP peptides, suggesting that production of biologically relevant CTL escape virus variants in vivo requires selection of mutations in more than one and likely all the CTL epitopes, a low probability event.


Asunto(s)
Antígenos H-2/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Conformación Proteica , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/virología , Proteínas Virales/inmunología , Replicación Viral , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Células Clonales , Simulación por Computador , Cartilla de ADN , Variación Genética , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/síntesis química , Péptidos/química , Péptidos/inmunología , Fenotipo , Mutación Puntual , Bazo/inmunología , Proteínas Virales/biosíntesis , Proteínas Virales/química
15.
J Virol ; 69(2): 1059-70, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7815484

RESUMEN

Despite the clinical importance of virus-induced immunosuppression, how virus infection may lead to a generalized suppression of the host immune response is poorly understood. To elucidate the principles involved, we analyzed the mechanism by which a lymphocytic choriomeningitis virus (LCMV) variant produces a generalized immune suppression in its natural host, the mouse. Whereas adult mice inoculated intravenously with LCMV Armstrong rapidly clear the infection and remain immunocompetent, inoculation with the Armstrong-derived LCMV variant clone 13, which differs from its parent virus at only two amino acid positions, by contrast results in persistent infection and a generalized deficit in responsiveness to subsequent immune challenge. Here we show that the immune suppression induced by LCMV clone 13 is associated with a CD8-dependent loss of interdigitating dendritic cells from periarteriolar lymphoid sheaths in the spleen and, functionally, with a deficit in the ability of splenocytes from infected mice to stimulate the proliferation of naive T cells in a primary mixed lymphocyte reaction. Dendritic cells are not depleted in immunocompetent Armstrong-infected mice. LCMV Armstrong and clone 13 exhibit differences in their tropism within the spleen, with clone 13 causing a higher level of infection of antigen-presenting cells in the white pulp, including periarterial interdigitating dendritic cells, than Armstrong, thereby rendering these cells targets for destruction by the antiviral CD8+ cytotoxic T-lymphocyte response which is induced at early times following infection with either virus. Our findings illustrate the key role that virus tropism may play in determining pathogenicity and, further, document a mechanism for virus-induced immunosuppression which may contribute to the clinically important immune suppression associated with many virus infections, including human immunodeficiency virus type 1.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica , Coriomeningitis Linfocítica/inmunología , Animales , Células Presentadoras de Antígenos/fisiología , Antígenos CD8/fisiología , Cricetinae , Infecciones por VIH/inmunología , Ratones , Ratones Endogámicos BALB C , Bazo/virología , Linfocitos T Citotóxicos/inmunología
16.
J Virol ; 68(11): 7367-73, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7933120

RESUMEN

Infection of neonatal mice with lymphocytic choriomeningitis virus (LCMV) strain Armstrong (ARM) results in a lifelong persistent infection. Viral variants (cytotoxic T lymphocyte [CTL] negative, persistence positive [CTL- P+]) can be isolated from the lymphoid tissues of such mice. Adult mice inoculated with these CTL- P+ viruses fail to generate sufficient cytotoxic T lymphocytes to clear the acute infection and become persistently infected. By contrast, inoculation of a similar dose of the parental ARM virus (CTL+ P-) into adult mice leads to the generation of a vigorous virus-specific CTL response that clears the infection. Sequence analysis revealed a phenylalanine (Phe)-to-Leucine (Leu) change at amino acid 260 of the viral glycoprotein (GP) as a marker for variant viruses with the CTL- P+ phenotype. An RNA PCR assay that detects the variant GP sequence and thus allows kinetic studies of the selection of the Leu at position 260 was developed. We found that although CTL- P+ viruses are known to be lymphotropic, mature T and B cells were not required for the generation and selection of the Leu at GP amino acid 260. Kinetically, in mice infected at birth with LCMV ARM, as early as 3 weeks postinfection the Phe-to-Leu change was found in virus in the serum. By 5 weeks, viral nucleic acid obtained from peritoneal macrophages, spleen, lymph nodes, and liver showed the Phe-to-Leu change. At 2 months postinfection, the Leu change was detected in virus from the thymus, heart, lung, and kidney. By contrast, virus replicating in the central nervous system showed only minimal levels of the Leu change by 4 months and as long as 1 year postinfection. In vitro studies showed that the parental LCMV ARM CTL+ P- virus replicates more efficiently and outcompetes CTL- P+ virus in a cultured neuronal cell line, indicating that differential growth properties in neurons are likely the basis for the selection of the parental virus over the CTL- P+ variant in the brain.


Asunto(s)
Tolerancia Inmunológica , Virus de la Coriomeningitis Linfocítica/genética , Mutación , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Glicoproteínas/genética , Cinética , Virus de la Coriomeningitis Linfocítica/crecimiento & desarrollo , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Datos de Secuencia Molecular , Neuronas/virología , Reacción en Cadena de la Polimerasa , ARN Viral/química , Linfocitos T Citotóxicos/inmunología
17.
J Neuroimmunol ; 51(2): 147-52, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-8182114

RESUMEN

The invariably fatal immunopathological disease that follows intracerebral injection of CBA/Ca (H-2k) mice with 1000 PFU of lymphocytic choriomeningitis virus (LCMV) generally fails to develop in congenic mice transgenic for a V beta 8.1D beta 2J beta 2.3C beta 2 T cell receptor (TCR) gene. The majority of these LCMV-infected TCR-transgenic mice show a substantial meningitis of delayed onset, that resolves without causing any obvious clinical impairment. This inflammatory process depends on the involvement of V beta 8+ T cells, but does not require the participation of the CD4+ subset. The cytotoxic effectors that develop in both the transgenic mice and the CBA/Ca controls are lytic for target cells infected with a vaccinia construct expressing genes encoding the putative polymerase protein of LCMV. Limiting the available TCR repertoire to lymphocytes with a single V beta phenotype (not required for the generation of potent effectors in wild-type mice) thus modifies the development of the lethal neuropathology characteristic of LCMV infection, although the CD8+ cytotoxic T lymphocyte response is not greatly compromised.


Asunto(s)
Coriomeningitis Linfocítica/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/fisiología , Enfermedad Aguda , Animales , Femenino , Coriomeningitis Linfocítica/etiología , Coriomeningitis Linfocítica/patología , Masculino , Ratones , Ratones Endogámicos CBA , Ratones Transgénicos , Linfocitos T Citotóxicos/inmunología
18.
J Immunol ; 151(11): 6259-64, 1993 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7902375

RESUMEN

The major host response to many viral infections is the generation of virus-specific CTL. Many protein molecules on the surfaces of both CTL and target cells interact to mediate adhesion of the cells and generate signals that lead to T cell activation and proliferation of virus-specific CTL that then mediate lysis of infected cells. One such protein, CD2, has been shown to increase the binding affinity of CTL to infected cells, and, in addition, enhance CTL activation signals. To determine whether virus-specific CTL could be generated in the absence of CD2, mice lacking a functional CD2 gene were infected with lymphocytic choriomeningitis virus (LCMV), and the responses to the virus were monitored. CD2-deficient mice infected intracerebrally with LCMV died as a consequence of CTL-mediated choriomeningitis, similar to control littermates. Additionally, CD2-deficient mice inoculated i.p. with LCMV cleared the infection by 2 wk postinfection, as did control mice. Viral clearance in these mice was shown to be due to the generation of a vigorous virus-specific MHC-restricted CTL response. Finally, to determine whether CD2 is essential for the generation of memory CTL, we examined the ability of CD2-deficient mice to generate memory CTL to LCMV and found normal memory CTL responses. Our results indicate that CD2 is not required for the generation of an LCMV-specific CTL response in vivo, nor is CD2 required for the maintenance or activation of memory CTL.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/fisiología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores Inmunológicos/deficiencia , Linfocitos T Citotóxicos/fisiología , Animales , Antígenos CD2 , Femenino , Complejo Mayor de Histocompatibilidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Receptores Inmunológicos/fisiología
19.
Proc Natl Acad Sci U S A ; 88(16): 6986-90, 1991 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-1871114

RESUMEN

The RNA subunit of Saccharomyces cerevisiae nuclear RNase P is encoded by a single-copy, essential gene, RPR1. The 369-nucleotide mature form of the RNA has an apparent precursor with an 84-nucleotide 5' leader and approximately 33 nucleotides of additional 3' sequence. Analysis of RPR1 transcription in a strain with a temperature-sensitive lesion in RNA polymerase III shows that the gene is transcribed in vivo by RNA polymerase III. Examination of potential promoter regions using both progressive upstream deletions and point mutations indicates that at least two sequences contained within the 5' leader region are essential for expression in vivo, while sequences farther upstream influence efficiency. The required leader elements resemble tRNA gene-like A-box and B-box internal promoters in sequence and spacing. As in the tRNA genes, transcription factor TFIIIC binds to this region in vitro and binding is severely reduced by either A-box or B-box point mutations that impair expression in vivo. It thus appears that the yeast RNase P RNA gene has adopted a promoter strategy that places an RNA polymerase III "internal" promoter upstream of the mature structural domain to help drive transcription.


Asunto(s)
Endorribonucleasas/genética , Regulación Fúngica de la Expresión Génica , Genes Fúngicos , Regiones Promotoras Genéticas , ARN Polimerasa III/genética , ARN Catalítico/genética , ARN de Hongos/genética , Saccharomyces cerevisiae/genética , Factores de Transcripción TFIII , Secuencia de Bases , ADN de Hongos/genética , ADN de Hongos/metabolismo , Prueba de Complementación Genética , Sustancias Macromoleculares , Datos de Secuencia Molecular , Ribonucleasa P , Saccharomyces cerevisiae/enzimología , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/metabolismo
20.
Biochem Biophys Res Commun ; 176(2): 826-32, 1991 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-2025294

RESUMEN

Complexes between transcription factor TFIIIC and eukaryotic tRNA gene internal promoter A and B boxes are unusual in that the binding to the two distinct sites tolerates considerable variation in both distance and helical orientation between the sites. Electrophoretic mobility of Saccharomyces cerevisiae TFIIIC complexes with circularly permuted tRNA gene fragments and sensitivity of the complexes to a single stranded-specific reagent, potassium permanganate, indicated that no significant bend or distortion was introduced into the DNA by simultaneous binding to both internal promoters. These data support a model in which variability in the relative positions of the two binding sites is compensated by flexibility in the structure of TFIIIC.


Asunto(s)
Regiones Promotoras Genéticas , ARN de Transferencia/metabolismo , Factores de Transcripción TFIII , Factores de Transcripción/genética , ADN/metabolismo , Desoxirribonucleasa I , Mapeo Restrictivo , Saccharomyces cerevisiae/análisis , Sensibilidad y Especificidad , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...