Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Am J Transplant ; 20(2): 399-410, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31595669

RESUMEN

Donor-specific antibodies (DSAs) have a deleterious effect on allografts and remain a major immunologic barrier in transplantation. Current therapies to eliminate DSAs are ineffective in highly HLA-sensitized patients. Proteasome inhibitors have been employed as a strategy to target bone marrow plasma cells (BMPCs), the source of long-term antibody production; however, their efficacy has been limited by poorly defined drug-resistance mechanisms. Here, we performed transcriptomic profiling of CD138+ BMPCs that survived in vivo desensitization therapy with the proteasome inhibitor carfilzomib to identify mechanisms of drug resistance. The results revealed a genomic signature that included increased expression of the immunoproteasome, a highly specialized proteasomal variant. Western blotting and functional studies demonstrated that catalytically active immunoproteasomes and the immunoproteasome activator PA28 were upregulated in carfilzomib-resistant BMPCs. Carfilzomib-resistant BMPCs displayed reduced sensitivity to the proteasome inhibitors carfilzomib, bortezomib, and ixazomib, but enhanced sensitivity to an immunoproteasome-specific inhibitor ONX-0914. Finally, in vitro carfilzomib treatment of BMPCs from HLA-sensitized patients increased levels of the immunoproteasome ß5i (PSMB8) catalytic subunit suggesting that carfilzomib therapy directly induces an adaptive immunoproteasome response. Taken together, our results indicate that carfilzomib induces structural changes in proteasomes and immunoproteasome formation.


Asunto(s)
Médula Ósea/efectos de los fármacos , Resistencia a Medicamentos/genética , Oligopéptidos/farmacología , Células Plasmáticas/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Transcriptoma/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/inmunología , Biomarcadores/metabolismo , Western Blotting , Médula Ósea/inmunología , Humanos , Células Plasmáticas/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Sindecano-1/metabolismo , Transcriptoma/inmunología , Investigación Biomédica Traslacional , Regulación hacia Arriba
2.
Am J Hum Genet ; 100(4): 676-688, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28343629

RESUMEN

Ubiquitination is a posttranslational modification that regulates many cellular processes including protein degradation, intracellular trafficking, cell signaling, and protein-protein interactions. Deubiquitinating enzymes (DUBs), which reverse the process of ubiquitination, are important regulators of the ubiquitin system. OTUD6B encodes a member of the ovarian tumor domain (OTU)-containing subfamily of deubiquitinating enzymes. Herein, we report biallelic pathogenic variants in OTUD6B in 12 individuals from 6 independent families with an intellectual disability syndrome associated with seizures and dysmorphic features. In subjects with predicted loss-of-function alleles, additional features include global developmental delay, microcephaly, absent speech, hypotonia, growth retardation with prenatal onset, feeding difficulties, structural brain abnormalities, congenital malformations including congenital heart disease, and musculoskeletal features. Homozygous Otud6b knockout mice were subviable, smaller in size, and had congenital heart defects, consistent with the severity of loss-of-function variants in humans. Analysis of peripheral blood mononuclear cells from an affected subject showed reduced incorporation of 19S subunits into 26S proteasomes, decreased chymotrypsin-like activity, and accumulation of ubiquitin-protein conjugates. Our findings suggest a role for OTUD6B in proteasome function, establish that defective OTUD6B function underlies a multisystemic human disorder, and provide additional evidence for the emerging relationship between the ubiquitin system and human disease.


Asunto(s)
Anomalías Múltiples/genética , Endopeptidasas/genética , Discapacidad Intelectual/genética , Adolescente , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Linaje , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Convulsiones/genética
3.
J Biol Chem ; 291(16): 8805-15, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26903513

RESUMEN

The supply of MHC class I-restricted peptides is primarily ensured by the degradation of intracellular proteins via the ubiquitin-proteasome system. Depending on the target and the enzymes involved, ubiquitination is a process that may dramatically vary in terms of linkages, length, and attachment sites. Here we identified the unique lysine residue at position 124 of the NY-ESO-1 cancer/testis antigen as the acceptor site for the formation of canonical Lys-48-linkages. Interestingly, a lysine-less form of NY-ESO-1 was as efficient as its wild-type counterpart in supplying the HLA-A*0201-restricted NY-ESO-1157-165 antigenic peptide. In fact, we show that the regulation of NY-ESO-1 processing by the ubiquitin receptors Rpn10 and Rpn13 as a well as by the standard and immunoproteasome is governed by non-canonical ubiquitination on non-lysine sites. In summary, our data underscore the significance of atypical ubiquitination in the modulation of MHC class I antigen processing.


Asunto(s)
Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígeno HLA-A2/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de la Membrana/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Ubiquitinación/inmunología , Antígenos de Neoplasias/genética , Antígeno HLA-A2/genética , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Complejo de la Endopetidasa Proteasomal/genética , Proteínas de Unión al ARN , Ubiquitinación/genética
4.
Eur J Immunol ; 45(12): 3257-68, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26399368

RESUMEN

The immunodominant MART-1(26(27)-35) epitope, liberated from the differentiation antigen melanoma antigen recognized by T cells/melanoma antigen A (MART-1/Melan-A), has been frequently targeted in melanoma immunotherapy, but with limited clinical success. Previous studies suggested that this is in part due to an insufficient peptide supply and epitope presentation, since proteasomes containing the immunosubunits ß5i/LMP7 (LMP, low molecular weight protein) or ß1i/LMP2 and ß5i/LMP7 interfere with MART-1(26-35) epitope generation in tumor cells. Here, we demonstrate that in addition the IFN-γ-inducible proteasome subunit ß2i/MECL-1 (multicatalytic endopeptidase complex-like 1), proteasome activator 28 (PA28), and ER-resident aminopeptidase 1 (ERAP1) impair MART-1(26-35) epitope generation. ß2i/MECL-1 and PA28 negatively affect C- and N-terminal cleavage and therefore epitope liberation from the proteasome, whereas ERAP1 destroys the MART-1(26-35) epitope by overtrimming activity. Constitutive expression of PA28 and ERAP1 in melanoma cells indicate that both interfere with MART-1(26-35) epitope generation even in the absence of IFN-γ. In summary, our results provide first evidence that activities of different antigen-processing components contribute to an inefficient MART-1(26-35) epitope presentation, suggesting the tumor cell's proteolytic machinery might have an important impact on the outcome of epitope-specific immunotherapies.


Asunto(s)
Aminopeptidasas/fisiología , Epítopos/inmunología , Melanoma/inmunología , Proteínas Musculares/fisiología , Proteínas de Neoplasias/inmunología , Complejo de la Endopetidasa Proteasomal/fisiología , Linfocitos T/inmunología , Línea Celular Tumoral , Cisteína Endopeptidasas/fisiología , Humanos , Antígenos de Histocompatibilidad Menor
5.
Apoptosis ; 20(9): 1211-28, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26201457

RESUMEN

Proteasome activator PA28γ (REGγ, Ki antigen) has recently been demonstrated to display anti-apoptotic properties via enhancing Mdm2-p53 interaction, thereby facilitating ubiquitination and down-regulation of the tumor suppressor p53. In this study we demonstrate a correlation between cellular PA28γ levels and the sensitivity of cells towards apoptosis in different cellular contexts thereby confirming a role of proteasome activator PA28γ as an anti-apoptotic regulator. We investigated the anti-apoptotic role of PA28γ upon UV-C stimulation in B8 mouse fibroblasts stably overexpressing the PA28γ-encoding PSME3 gene and upon butyrate-induced apoptosis in human HT29 adenocarcinoma cells with silenced PSME3 gene. Interestingly, our results demonstrate that PA28γ has a strong influence on different apoptotic hallmarks, especially p53 phosphorylation and caspase activation. In detail, PA28γ and effector caspases mutually restrict each other. PA28γ is a caspase substrate, if PA28γ levels are low. In contrast, PA28γ overexpression reduces caspase activities, including the caspase-dependent processing of PA28γ. Furthermore, overexpression of PA28γ resulted in a nuclear accumulation of transcriptional active p53. In summary, our findings indicate that even in a p53-dominated cellular context, pro-apoptotic signaling might be overcome by PA28γ-mediated caspase inhibition.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autoantígenos/metabolismo , Caspasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Apoptosis/efectos de la radiación , Butiratos/farmacología , Citocromos c/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de la radiación , Células HT29/efectos de la radiación , Humanos , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Rayos Ultravioleta/efectos adversos
6.
J Cell Mol Med ; 18(1): 59-68, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24304442

RESUMEN

The 20S proteasome is almost exclusively localized within cells. High levels of extracellular proteasomes are also found circulating in the blood plasma of patients suffering from a variety of inflammatory, autoimmune and neoplastic diseases. However, the origin of these proteasomes remained enigmatic. Since the proteome of microparticles, small membrane enclosed vesicles released from cells, was shown to contain proteasomal subunits, we studied whether intact proteasomes are actively released into the extracellular space. Using human primary T lymphocytes stimulated with CaCl2 and the calcium ionophore A23187 to induce membrane blebbing we demonstrate that microparticles contain proteolytically active 20S proteasomes as well as the proteasome activator PA28 and subunits of the 19S proteasome regulator. Furthermore, our experiments reveal that incubation of in vitro generated T lymphocyte-microparticles with sphingomyelinase results in the hydrolysis of the microparticle membranes and subsequent release of proteasomes from the vesicles. Thus, we here show for the first time that functional proteasomes can be exported from activated immune cells by way of microparticles, the dissolution of which may finally lead to the generation of extracellular proteasomes.


Asunto(s)
Micropartículas Derivadas de Células/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Linfocitos T/metabolismo , Células Cultivadas , Humanos , Subunidades de Proteína/metabolismo , Esfingomielina Fosfodiesterasa/química , Linfocitos T/enzimología
7.
J Exp Biol ; 217(Pt 19): 3441-6, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063852

RESUMEN

In honeybees (Apis mellifera), the proteasome inhibitor Z-Leu-Leu-Leu-CHO (MG132) enhances long-term memory (LTM) formation. Studies in vertebrates using different inhibitors of the proteasome demonstrate the opposite, namely an inhibition of memory formation. The reason for this contradiction remains unclear. MG132 is an inhibitor of the proteasome, but also blocks other proteases. Accordingly, one possible explanation might be that other proteases affected by MG132 are responsible for the enhancement of LTM formation. We test this hypothesis by comparing the effect of MG132 and the more specific proteasome inhibitor clasto-lactacystin beta-lactone (ß-lactone). We show that these two inhibitors block the activity of the proteasome in honeybee brains to a similar extent, do not affect the animals' survival but do enhance LTM retention upon olfactory conditioning. Thus, the enhancement of LTM formation is not due to MG132-specific side effects, but to inhibition of a protease targeted by MG132 and ß-lactone, i.e. the proteasome.


Asunto(s)
Abejas/fisiología , Condicionamiento Clásico/fisiología , Memoria a Largo Plazo/fisiología , Inhibidores de Proteasoma/farmacología , Ubiquitina/metabolismo , Animales , Abejas/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Lactonas/metabolismo , Lactonas/farmacología , Leupeptinas/metabolismo , Leupeptinas/farmacología , Memoria a Largo Plazo/efectos de los fármacos , Odorantes , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Complejo de la Endopetidasa Proteasomal/fisiología
8.
Mol Cell Proteomics ; 11(8): 467-77, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22556278

RESUMEN

The post-translational modification of proteins with O-GlcNAc is involved in various cellular processes including signal transduction, transcription, translation, and nuclear transport. This transient protein modification enables cells or tissues to adapt to nutrient conditions or stress. O-Glycosylation of the 26 S proteasome ATPase subunit Rpt2 is known to influence the stability of proteins by reducing their proteasome-dependent degradation. In contrast, knowledge of the sites of O-GlcNAcylation on the subunits of the catalytic core of the 26 S proteasome, the 20 S proteasome, and the impact on proteasome activity is very limited. This is predominantly because O-GlcNAc modifications are often substoichiometric and because 20 S proteasomes represent a complex protein mixture of different subtypes. Therefore, identification of O-GlcNAcylation sites on proteasome subunits essentially requires effective enrichment strategies. Here we describe an adapted ß-elimination-based derivatization method of O-GlcNAc peptides using a novel biotin-cystamine tag. The specificity of the reaction was increased by differential isotopic labeling with either "light" biotin-cystamine or deuterated "heavy" biotin-cystamine. The enriched peptides were analyzed by LC-MALDI-TOF/TOF-MS and relatively quantified. The method was optimized using bovine α-crystallin and then applied to murine 20 S proteasomes isolated from spleen and brain and murine Hsp90 isolated from liver. Using this approach, we identified five novel and one known O-GlcNAc sites within the murine 20 S proteasome core complex that are located on five different subunits and in addition two novel O-GlcNAc sites on murine Hsp90ß, of which one corresponds to a previously described phosphorylation site.


Asunto(s)
Acetilglucosamina/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Biotina/metabolismo , Western Blotting , Radioisótopos de Carbono/metabolismo , Bovinos , Cromatografía Liquida , Cistamina/metabolismo , Glucosa/farmacología , Glicosilación/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , alfa-Cristalinas/metabolismo
9.
Mol Cell Proteomics ; 11(10): 1008-23, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22822185

RESUMEN

Proteasome-catalyzed peptide splicing (PCPS) represents an additional activity of mammalian 20S proteasomes recently identified in connection with antigen presentation. We show here that PCPS is not restricted to mammalians but that it is also a feature of yeast 20S proteasomes catalyzed by all three active site ß subunits. No major differences in splicing efficiency exist between human 20S standard- and immuno-proteasome or yeast 20S proteasome. Using H(2)(18)O to monitor the splicing reaction we also demonstrate that PCPS occurs via direct transpeptidation that slightly favors the generation of peptides spliced in cis over peptides spliced in trans. Splicing efficiency itself is shown to be controlled by proteasomal cleavage site preference as well as by the sequence characteristics of the spliced peptides. By use of kinetic data and quantitative analyses of PCPS obtained by mass spectrometry we developed a structural model with two PCPS binding sites in the neighborhood of the active Thr1.


Asunto(s)
Linfocitos B/metabolismo , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Empalme de Proteína , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Linfocitos B/citología , Biocatálisis , Línea Celular Transformada , Cromatografía Liquida , Humanos , Datos de Secuencia Molecular , Péptidos/síntesis química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
Ann Hepatol ; 13(4): 429-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24927614

RESUMEN

BACKGROUND: The 20S proteasome is the proteolytic core of the major intracellular protein degradative system, the ubiquitin-proteasome system. Since little is known about proteasomes of human liver, we have investigated the proteasome spectrum in adult human liver. MATERIAL AND METHODS: 20S proteasomes were chromatographically purified from adult human liver and from HuH7 cells. They were divided into subpopulations and subtypes and characterized with regard to their proteolytic activities using short fluorogenic oligo- and long poly-peptide substrates. Their subunit composition was studied by immunoblotting. RESULTS: Proteasomes from adult human liver tissue can be separated into three subpopulations (I, II, III), each of which is composed of several subtypes, which total to a spectrum of 14 different subtypes. Two minor subtypes contain only the immuno-subunits ß1i and ß5i but not their standard counterparts; all others are intermediate subtypes containing ß1 and ß5 standard- and ß1i and ß5i immuno-subunits in various compositions. With regard to the proteolytic activities we observed that a decreasing content of subunit ß1i in the subtypes goes along with a decreasing ratio of chymotrypsin-like/caspase-like activity, whereas the degradation rate of a 30 mer polypeptide substrate increased with decreasing ß1i content. By comparison, 20S proteasomes from HuH7 cells do not contain immuno-subunits but are pure standard proteasomes, which can be separated into three subtypes. CONCLUSION: These findings suggest that adult human liver contains a spectrum of 14 different 20S proteasome subtypes with different enzymatic properties reflecting most probably an adaptive response of liver cell functions to challenging factors during lifetime.


Asunto(s)
Eritrocitos/enzimología , Hígado/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Bazo/enzimología , Línea Celular , Electroforesis , Humanos
11.
Blood ; 118(4): 946-54, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21613253

RESUMEN

Adoptive therapy with T-cell receptor (TCR)-engineered T cells is a promising approach in cancer treatment. While usage of T cells specific for tumor-associated antigens (TAAs) can lead to serious side effects because of autoimmunity, targeting true tumor-specific mutations, such as the products of translocations in leukemias, should reduce such a risk. A potentially ideal target might be the chimeric protein TEL-AML1, which results from the chromosomal translocation 12;21 and represents the most common fusion gene in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Within the fusion region of TEL-AML1, a single epitope has been described by reverse immunology as immunogenic in HLA-A*0201 restriction settings. As a potential source of TCRs specific for this TEL-AML1 epitope, we have used mice expressing a human TCR-αß repertoire and human MHC class I. Surprisingly, we have found that, although a specific functional CD8(+) T-cell response against this peptide could be evoked, the described epitope was in fact not endogenously processed. Analyses done with a potent antigen-presenting cell line, as well as with purified human proteasomes, support the conclusion that this peptide cannot be proposed as a potential target in immunotherapy of ALL in HLA-A*0201-restricted fashion.


Asunto(s)
Presentación de Antígeno/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Epítopos de Linfocito T/inmunología , Proteínas de Fusión Oncogénica/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Separación Celular , Cromatografía Líquida de Alta Presión , Técnicas de Cocultivo , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Translocación Genética
12.
Cell Mol Life Sci ; 69(14): 2443-54, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22349260

RESUMEN

Like ubiquitin (Ub), the ubiquitin-like protein FAT10 can serve as a signal for proteasome-dependent protein degradation. Here, we investigated the contribution of FAT10 substrate modification to MHC class I antigen presentation. We show that N-terminal modification of the human cytomegalovirus-derived pp65 antigen to FAT10 facilitates direct presentation and dendritic cell-mediated cross-presentation of the HLA-A2 restricted pp65(495-503) epitope. Interestingly, our data indicate that the pp65 presentation initiated by either FAT10 or Ub partially relied on the 19S proteasome subunit Rpn10 (S5a). However, FAT10 distinguished itself from Ub in that it promoted a pp65 response which was not influenced by immunoproteasomes or PA28. Further divergence occurred at the level of Ub-binding proteins with NUB1 supporting the pp65 presentation arising from FAT10, while it exerted no effect on that initiated by Ub. Collectively, our data establish FAT10 modification as a distinct and alternative signal for facilitated MHC class I antigen presentation.


Asunto(s)
Presentación de Antígeno , Antígenos de Histocompatibilidad Clase I/metabolismo , Ubiquitina/metabolismo , Ubiquitinas/metabolismo , Citomegalovirus/metabolismo , Células HEK293 , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Células HeLa , Humanos , Proteínas Musculares/antagonistas & inhibidores , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Proteolisis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo
13.
Cell Mol Life Sci ; 69(15): 2543-58, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22382925

RESUMEN

The proteasome is a multi-catalytic protein complex whose primary function is the degradation of abnormal or foreign proteins. Upon exposure of cells to interferons (IFNs), the ß1i/LMP2, ß2i/MECL-1, and ß5i/LMP7 subunits are induced and incorporated into newly synthesized immunoproteasomes (IP), which are thought to function solely as critical players in the optimization of the CD8(+) T-cell response. However, the observation that IP are present in several non-immune tissues under normal conditions and/or following pathological events militates against the view that its role is limited to MHC class I presentation. In support of this concept, the recent use of genetic models deficient for ß1i/LMP2, ß2i/MECL-1, or ß5i/LMP7 has uncovered unanticipated functions for IP in innate immunity and non-immune processes. Herein, we review recent data in an attempt to clarify the role of IP beyond MHC class I epitope presentation with emphasis on its involvement in the regulation of protein homeostasis, cell proliferation, and cytokine gene expression.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/inmunología , Complejo de la Endopetidasa Proteasomal/fisiología , Animales , Presentación de Antígeno , Proliferación Celular , Citocinas/biosíntesis , Citocinas/genética , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/metabolismo , Homeostasis , Humanos , Inmunidad Innata , Inflamación/inmunología , Inflamación/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Proteínas/metabolismo , Transducción de Señal , Estrés Fisiológico
14.
Mol Immunol ; 154: 61-68, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36608422

RESUMEN

The therapy of cancer by adoptive T cell transfer (ACT) requires T cell receptors (TCRs) with optimal affinity for HLA class I-bound peptides (pHLA-I). But not every patient responds to ACT. Therefore, it is critical to understand the individual factors influencing the recognition of HLA class I-bound peptides (pHLA-I) by TCRs. Focusing on three immunotherapy-targeted human HLA-A* 02:01-presented T cell epitopes we investigated the contribution of the ER-resident aminopeptidases ERAP1 and ERAP2 to TCR recognition of cancer cells. We found that ERAP2 on its own, when expressed in ERAP-deficient cells, elicited a strong CTL response towards the Tyrosinase368-376 epitope. In vitro generated TAP-dependent N-terminally extended epitope precursor peptides were differently customized by ERAP1 and ERAP2 and thus may serve as potential source for the Tyrosinase368-376 epitope. ERAP2 also influenced recognition of the gp100209-217 tumor epitope and enhanced T cell recognition of the MART-126/27-35 epitope in the absence of ERAP1 expression. Our results underline the relevance of ERAP2 for tumor epitope presentation and TCR recognition and may need to be considered when designing ACT in the future.


Asunto(s)
Monofenol Monooxigenasa , Neoplasias , Humanos , Péptidos/metabolismo , Epítopos de Linfocito T , Receptores de Antígenos de Linfocitos T , Inmunoterapia , Aminopeptidasas , Antígenos de Histocompatibilidad Menor/metabolismo
15.
J Biol Chem ; 286(7): 5151-6, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21149444

RESUMEN

Accumulation of aberrant proteins in the endoplasmic reticulum (ER) triggers the unfolded protein response pathway that helps the cell to survive under these stress conditions. Herp is a mammalian ubiquitin domain protein, which is strongly induced by the unfolded protein response. It is involved in ER-associated protein degradation (ERAD) and interacts directly with the ubiquitin ligase Hrd1, which is found in high molecular mass complexes of the ER membrane. Here we present the first evidence that Herp regulates Hrd1-mediated ubiquitylation in a ubiquitin-like (UBL) domain-dependent manner. We found that upon exposure of cells to ER stress, elevation of Herp steady state levels is accompanied by an enhanced association of Herp with pre-existing Hrd1. Hrd1-associated Herp is rapidly degraded and substituted by de novo synthesized Herp, suggesting a continuous turnover of the protein at Hrd1 complexes. Further analysis revealed the presence of multiple Hrd1 copies in a single complex enabling binding of a variable number of Herp molecules. Efficient ubiquitylation of the Hrd1-specific ERAD substrate α1-antitrypsin null Hong Kong (NHK) required the presence of the Herp UBL domain, which was also necessary for NHK degradation. In summary, we propose that binding of Herp to Hrd1-containing ERAD complexes positively regulates the ubiquitylation activity of these complexes, thus permitting survival of the cell during ER stress.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Respuesta de Proteína Desplegada , Supervivencia Celular , Retículo Endoplásmico/genética , Células HeLa , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Estructura Terciaria de Proteína , Ubiquitina/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética
16.
Eur J Immunol ; 41(4): 926-35, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21360704

RESUMEN

Proteasomes play a fundamental role in the processing of intracellular antigens into peptides that bind to MHC class I molecules for the presentation of CD8(+) T cells. Three IFN-γ-inducible catalytic proteasome (immuno)subunits as well as the IFN-γ-inducible proteasome activator PA28 dramatically accelerate the generation of a subset of MHC class I-presented antigenic peptides. To determine whether these IFN-γ-inducible proteasome components play a compounded role in antigen processing, we generated mice lacking both PA28 and immunosubunits ß5i/LMP7 and ß2i/MECL-1. Analyses of MHC class I cell-surface levels ex vivo demonstrated that PA28 deficiency reduced the production of MHC class I-binding peptides both in cells with and without immunosubunits, in the latter cells further decreasing an already diminished production of MHC ligands in the absence of immunoproteasomes. In contrast, the immunosubunits but not PA28 appeared to be of critical importance for the induction of CD8(+) T-cell responses to multiple dominant Influenza and Listeria-derived epitopes. Taken together, our data demonstrate that PA28 and the proteasome immunosubunits use fundamentally different mechanisms to enhance the supply of MHC class I-binding peptides; however, only the immunosubunit-imposed effects on proteolytic epitope processing appear to have substantial influence on the specificity of pathogen-specific CD8(+) T-cell responses.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Péptidos/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Péptidos/química , Complejo de la Endopetidasa Proteasomal/deficiencia
17.
FEBS J ; 289(13): 3826-3838, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35066984

RESUMEN

RNA viruses in the Picornaviridae family express a large 250 kDa viral polyprotein that is processed by virus-encoded proteinases into mature functional proteins with specific functions for virus replication. One of these proteins is the highly conserved enteroviral transmembrane protein 3A that assists in reorganizing cellular membranes associated with the Golgi apparatus. Here, we studied the molecular properties of the Coxsackievirus B3 (CVB3) protein 3A with regard to its dimerization and its functional stability. By applying mutational analysis and biochemical characterization, we demonstrate that protein 3A forms DTT-sensitive disulfide-linked dimers via a conserved cytosolic cysteine residue at position 38 (Cys38). Homodimerization of CVB3 protein 3A via Cys38 leads to profound stabilization of the protein, whereas a C38A mutation promotes a rapid proteasome-dependent elimination of its monomeric form. The lysosomotropic agent chloroquine (CQ) exerted only minor stabilizing effects on the 3A monomer but resulted in enrichment of the homodimer. Our experimental data demonstrate that disulfide linkages via a highly conserved Cys-residue in enteroviral protein 3A have an important role in the dimerization of this viral protein, thereby preserving its stability and functional integrity.


Asunto(s)
Disulfuros , Enterovirus Humano B , Dimerización , Disulfuros/metabolismo , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , Células HeLa , Humanos , Proteínas Virales/metabolismo , Replicación Viral
18.
Viruses ; 14(4)2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35458499

RESUMEN

Enteroviruses (EV) are implicated in an extensive range of clinical manifestations, such as pancreatic failure, cardiovascular disease, hepatitis, and meningoencephalitis. We recently reported on the biochemical properties of the highly conserved cysteine residue at position 38 (C38) of enteroviral protein 3A and demonstrated a C38-mediated homodimerization of the Coxsackievirus B3 protein 3A (CVB3-3A) that resulted in its profound stabilization. Here, we show that residue C38 of protein 3A supports the replication of CVB3, a clinically relevant member of the enterovirus genus. The infection of HeLa cells with protein 3A cysteine 38 to alanine mutants (C38A) attenuates virus replication, resulting in comparably lower virus particle formation. Consistently, in a mouse infection model, the enhanced virus propagation of CVB3-3A wt in comparison to the CVB3-3A[C38A] mutant was confirmed and found to promote severe liver tissue damage. In contrast, infection with the CVB3-3A[C38A] mutant mitigated hepatic tissue injury and ameliorated the signs of systemic inflammatory responses, such as hypoglycemia and hypothermia. Based on these data and our previous report on the C38-mediated stabilization of the CVB3-3A protein, we conclude that the highly conserved amino acid C38 in protein 3A enhances the virulence of CVB3.


Asunto(s)
Infecciones por Coxsackievirus , Infecciones por Enterovirus , Enterovirus , Animales , Cisteína , Enterovirus Humano B/fisiología , Células HeLa , Humanos , Ratones , Virulencia , Replicación Viral
19.
PLoS Comput Biol ; 6(6): e1000830, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20613855

RESUMEN

The identification of proteasome-generated spliced peptides (PSP) revealed a new unpredicted activity of the major cellular protease. However, so far characterization of PSP was entirely dependent on the availability of patient-derived cytotoxic CD8+ T lymphocytes (CTL) thus preventing a systematic investigation of proteasome-catalyzed peptide splicing (PCPS). For an unrestricted PSP identification we here developed SpliceMet, combining the computer-based algorithm ProteaJ with in vitro proteasomal degradation assays and mass spectrometry. By applying SpliceMet for the analysis of proteasomal processing products of four different substrate polypeptides, derived from human tumor as well as viral antigens, we identified fifteen new spliced peptides generated by PCPS either by cis or from two separate substrate molecules, i.e., by trans splicing. Our data suggest that 20S proteasomes represent a molecular machine that, due to its catalytic and structural properties, facilitates the generation of spliced peptides, thereby providing a pool of qualitatively new peptides from which functionally relevant products may be selected.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Fragmentos de Péptidos/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Programas Informáticos , Secuencia de Aminoácidos , Antígenos Nucleares/química , Antígenos Nucleares/metabolismo , Autoantígenos/química , Autoantígenos/metabolismo , Linfocitos T CD8-positivos , Simulación por Computador , Bases de Datos de Proteínas , Epítopos de Linfocito T , Factor 5 de Crecimiento de Fibroblastos/química , Factor 5 de Crecimiento de Fibroblastos/metabolismo , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Antígeno gp100 del Melanoma
20.
Viruses ; 13(7)2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34372566

RESUMEN

Infection by RNA viruses causes extensive cellular reorganization, including hijacking of membranes to create membranous structures termed replication organelles, which support viral RNA synthesis and virion assembly. In this study, we show that infection with coxsackievirus B3 entails a profound impairment of the protein homeostasis at virus-utilized membranes, reflected by an accumulation of ubiquitinylated proteins, including K48-linked polyubiquitin conjugates, known to direct proteins to proteasomal degradation. The enrichment of membrane-bound ubiquitin conjugates is attributed to the presence of the non-structural viral proteins 2B and 3A, which are known to perturb membrane integrity and can cause an extensive rearrangement of cellular membranes. The locally increased abundance of ubiquitinylated proteins occurs without an increase of oxidatively damaged proteins. During the exponential phase of replication, the oxidative damage of membrane proteins is even diminished, an effect we attribute to the recruitment of glutathione, which is known to be required for the formation of infectious virus particles. Furthermore, we show that the proteasome contributes to the processing of viral precursor proteins. Taken together, we demonstrate how an infection with coxsackievirus B3 affects the cellular protein and redox homeostasis locally at the site of viral replication and virus assembly.


Asunto(s)
Enterovirus Humano B/metabolismo , Ubiquitinación/fisiología , Replicación Viral/fisiología , Citoplasma/metabolismo , Enterovirus Humano B/patogenicidad , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/fisiología , ARN Viral/genética , Ubiquitina/metabolismo , Proteínas Virales/metabolismo , Virión/metabolismo , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA