Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Hum Genet ; 111(7): 1352-1369, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38866022

RESUMEN

Primary proteasomopathies have recently emerged as a new class of rare early-onset neurodevelopmental disorders (NDDs) caused by pathogenic variants in the PSMB1, PSMC1, PSMC3, or PSMD12 proteasome genes. Proteasomes are large multi-subunit protein complexes that maintain cellular protein homeostasis by clearing ubiquitin-tagged damaged, misfolded, or unnecessary proteins. In this study, we have identified PSMD11 as an additional proteasome gene in which pathogenic variation is associated with an NDD-causing proteasomopathy. PSMD11 loss-of-function variants caused early-onset syndromic intellectual disability and neurodevelopmental delay with recurrent obesity in 10 unrelated children. Our findings demonstrate that the cognitive impairment observed in these individuals could be recapitulated in Drosophila melanogaster with depletion of the PMSD11 ortholog Rpn6, which compromised reversal learning. Our investigations in subject samples further revealed that PSMD11 loss of function resulted in impaired 26S proteasome assembly and the acquisition of a persistent type I interferon (IFN) gene signature, mediated by the integrated stress response (ISR) protein kinase R (PKR). In summary, these data identify PSMD11 as an additional member of the growing family of genes associated with neurodevelopmental proteasomopathies and provide insights into proteasomal biology in human health.


Asunto(s)
Drosophila melanogaster , Discapacidad Intelectual , Mutación con Pérdida de Función , Trastornos del Neurodesarrollo , Obesidad , Fenotipo , Complejo de la Endopetidasa Proteasomal , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Obesidad/genética , Animales , Masculino , Niño , Femenino , Drosophila melanogaster/genética , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Preescolar , Adolescente , Interferones/metabolismo , Interferones/genética
2.
Am J Hum Genet ; 109(2): 361-372, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35051358

RESUMEN

Nuclear deubiquitinase BAP1 (BRCA1-associated protein 1) is a core component of multiprotein complexes that promote transcription by reversing the ubiquitination of histone 2A (H2A). BAP1 is a tumor suppressor whose germline loss-of-function variants predispose to cancer. To our knowledge, there are very rare examples of different germline variants in the same gene causing either a neurodevelopmental disorder (NDD) or a tumor predisposition syndrome. Here, we report a series of 11 de novo germline heterozygous missense BAP1 variants associated with a rare syndromic NDD. Functional analysis showed that most of the variants cannot rescue the consequences of BAP1 inactivation, suggesting a loss-of-function mechanism. In T cells isolated from two affected children, H2A deubiquitination was impaired. In matching peripheral blood mononuclear cells, histone H3 K27 acetylation ChIP-seq indicated that these BAP1 variants induced genome-wide chromatin state alterations, with enrichment for regulatory regions surrounding genes of the ubiquitin-proteasome system (UPS). Altogether, these results define a clinical syndrome caused by rare germline missense BAP1 variants that alter chromatin remodeling through abnormal histone ubiquitination and lead to transcriptional dysregulation of developmental genes.


Asunto(s)
Proteína BRCA1/genética , Mutación de Línea Germinal , Mutación con Pérdida de Función , Mutación Missense , Trastornos del Neurodesarrollo/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Adolescente , Proteína BRCA1/inmunología , Niño , Preescolar , Cromatina/química , Cromatina/inmunología , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/inmunología , Familia , Femenino , Regulación de la Expresión Génica , Heterocigoto , Histonas/genética , Histonas/inmunología , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/inmunología , Humanos , Lactante , Masculino , Trastornos del Neurodesarrollo/inmunología , Trastornos del Neurodesarrollo/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Linfocitos T/inmunología , Linfocitos T/patología , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/inmunología , Ubiquitina/genética , Ubiquitina/inmunología , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina Tiolesterasa/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
3.
Am J Hum Genet ; 109(10): 1909-1922, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36044892

RESUMEN

The transmembrane protein TMEM147 has a dual function: first at the nuclear envelope, where it anchors lamin B receptor (LBR) to the inner membrane, and second at the endoplasmic reticulum (ER), where it facilitates the translation of nascent polypeptides within the ribosome-bound TMCO1 translocon complex. Through international data sharing, we identified 23 individuals from 15 unrelated families with bi-allelic TMEM147 loss-of-function variants, including splice-site, nonsense, frameshift, and missense variants. These affected children displayed congruent clinical features including coarse facies, developmental delay, intellectual disability, and behavioral problems. In silico structural analyses predicted disruptive consequences of the identified amino acid substitutions on translocon complex assembly and/or function, and in vitro analyses documented accelerated protein degradation via the autophagy-lysosomal-mediated pathway. Furthermore, TMEM147-deficient cells showed CKAP4 (CLIMP-63) and RTN4 (NOGO) upregulation with a concomitant reorientation of the ER, which was also witnessed in primary fibroblast cell culture. LBR mislocalization and nuclear segmentation was observed in primary fibroblast cells. Abnormal nuclear segmentation and chromatin compaction were also observed in approximately 20% of neutrophils, indicating the presence of a pseudo-Pelger-Huët anomaly. Finally, co-expression analysis revealed significant correlation with neurodevelopmental genes in the brain, further supporting a role of TMEM147 in neurodevelopment. Our findings provide clinical, genetic, and functional evidence that bi-allelic loss-of-function variants in TMEM147 cause syndromic intellectual disability due to ER-translocon and nuclear organization dysfunction.


Asunto(s)
Discapacidad Intelectual , Anomalías Musculoesqueléticas , Anomalía de Pelger-Huët , Núcleo Celular/genética , Niño , Cromatina , Humanos , Discapacidad Intelectual/genética , Pérdida de Heterocigocidad , Anomalía de Pelger-Huët/genética
4.
Genet Med ; 26(6): 101120, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38469793

RESUMEN

PURPOSE: Imbalances in protein homeostasis affect human brain development, with the ubiquitin-proteasome system (UPS) and autophagy playing crucial roles in neurodevelopmental disorders (NDD). This study explores the impact of biallelic USP14 variants on neurodevelopment, focusing on its role as a key hub connecting UPS and autophagy. METHODS: Here, we identified biallelic USP14 variants in 4 individuals from 3 unrelated families: 1 fetus, a newborn with a syndromic NDD and 2 siblings affected by a progressive neurological disease. Specifically, the 2 siblings from the latter family carried 2 compound heterozygous variants c.8T>C p.(Leu3Pro) and c.988C>T p.(Arg330∗), whereas the fetus had a homozygous frameshift c.899_902del p.(Lys300Serfs∗24) variant, and the newborn patient harbored a homozygous frameshift c.233_236del p.(Leu78Glnfs∗11) variant. Functional studies were conducted using sodium dodecyl-sulfate polyacrylamide gel electrophoresis, western blotting, and mass spectrometry analyses in both patient-derived and CRISPR-Cas9-generated cells. RESULTS: Our investigations indicated that the USP14 variants correlated with reduced N-terminal methionine excision, along with profound alterations in proteasome, autophagy, and mitophagy activities. CONCLUSION: Biallelic USP14 variants in NDD patients perturbed protein degradation pathways, potentially contributing to disorder etiology. Altered UPS, autophagy, and mitophagy activities underscore the intricate interplay, elucidating their significance in maintaining proper protein homeostasis during brain development.


Asunto(s)
Trastornos del Neurodesarrollo , Humanos , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Femenino , Masculino , Alelos , Autofagia/genética , Ubiquitina Tiolesterasa/genética , Recién Nacido , Complejo de la Endopetidasa Proteasomal/genética , Linaje , Homocigoto , Predisposición Genética a la Enfermedad , Mutación/genética
5.
Genet Med ; 24(9): 1941-1951, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35678782

RESUMEN

PURPOSE: WNK3 kinase (PRKWNK3) has been implicated in the development and function of the brain via its regulation of the cation-chloride cotransporters, but the role of WNK3 in human development is unknown. METHOD: We ascertained exome or genome sequences of individuals with rare familial or sporadic forms of intellectual disability (ID). RESULTS: We identified a total of 6 different maternally-inherited, hemizygous, 3 loss-of-function or 3 pathogenic missense variants (p.Pro204Arg, p.Leu300Ser, p.Glu607Val) in WNK3 in 14 male individuals from 6 unrelated families. Affected individuals had ID with variable presence of epilepsy and structural brain defects. WNK3 variants cosegregated with the disease in 3 different families with multiple affected individuals. This included 1 large family previously diagnosed with X-linked Prieto syndrome. WNK3 pathogenic missense variants localize to the catalytic domain and impede the inhibitory phosphorylation of the neuronal-specific chloride cotransporter KCC2 at threonine 1007, a site critically regulated during the development of synaptic inhibition. CONCLUSION: Pathogenic WNK3 variants cause a rare form of human X-linked ID with variable epilepsy and structural brain abnormalities and implicate impaired phospho-regulation of KCC2 as a pathogenic mechanism.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X , Proteínas Serina-Treonina Quinasas , Simportadores , Encéfalo/anomalías , Dominio Catalítico/genética , Hemicigoto , Humanos , Mutación con Pérdida de Función , Masculino , Herencia Materna/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Mutación Missense , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Simportadores/metabolismo
6.
Genet Med ; 24(1): 179-191, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34906456

RESUMEN

PURPOSE: Haploinsufficiency of PSMD12 has been reported in individuals with neurodevelopmental phenotypes, including developmental delay/intellectual disability (DD/ID), facial dysmorphism, and congenital malformations, defined as Stankiewicz-Isidor syndrome (STISS). Investigations showed that pathogenic variants in PSMD12 perturb intracellular protein homeostasis. Our objective was to further explore the clinical and molecular phenotypic spectrum of STISS. METHODS: We report 24 additional unrelated patients with STISS with various truncating single nucleotide variants or copy-number variant deletions involving PSMD12. We explore disease etiology by assessing patient cells and CRISPR/Cas9-engineered cell clones for various cellular pathways and inflammatory status. RESULTS: The expressivity of most clinical features in STISS is highly variable. In addition to previously reported DD/ID, speech delay, cardiac and renal anomalies, we also confirmed preaxial hand abnormalities as a feature of this syndrome. Of note, 2 patients also showed chilblains resembling signs observed in interferonopathy. Remarkably, our data show that STISS patient cells exhibit a profound remodeling of the mTORC1 and mitophagy pathways with an induction of type I interferon-stimulated genes. CONCLUSION: We refine the phenotype of STISS and show that it can be clinically recognizable and biochemically diagnosed by a type I interferon gene signature.


Asunto(s)
Discapacidad Intelectual , Trastornos del Desarrollo del Lenguaje , Anomalías Musculoesqueléticas , Haploinsuficiencia , Humanos , Discapacidad Intelectual/diagnóstico , Trastornos del Desarrollo del Lenguaje/genética , Anomalías Musculoesqueléticas/genética , Fenotipo
7.
Cancer Immunol Immunother ; 70(10): 3015-3030, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34120214

RESUMEN

Adoptive cell transfer (ACT) of tumor-specific T lymphocytes represents a relevant therapeutic strategy to treat metastatic melanoma patients. Ideal T-cells should combine tumor specificity and reactivity with survival in vivo, while avoiding autoimmune side effects. Here we report results from a Phase I/II clinical trial (NCT02424916, performed between 2015 and 2018) in which 6 metastatic HLA-A2 melanoma patients received autologous antigen-specific T-cells produced from PBMC, after peptide stimulation in vitro, followed by sorting with HLA-peptide multimers and amplification. Each patient received a combination of Melan-A and MELOE-1 polyclonal specific T-cells, whose specificity and anti-tumor reactivity were checked prior to injection, with subcutaneous IL-2. Transferred T-cells were also characterized in terms of functional avidity, diversity and phenotype and their blood persistence was evaluated. An increase in specific T-cells was detected in the blood of all patients at day 1 and progressively disappeared from day 7 onwards. No serious adverse events occurred after this ACT. Clinically, five patients progressed and one patient experienced a partial response following therapy. Melan-A and MELOE-1 specific T-cells infused to this patient were diverse, of high avidity, with a high proportion of T lymphocytes co-expressing PD-1 and TIGIT but few other exhaustion markers. In conclusion, we demonstrated the feasibility and safety of ACT with multimer-sorted Melan-A and MELOE-1 specific T cells to metastatic melanoma patients. The clinical efficacy of such therapeutic strategy could be further enhanced by the selection of highly reactive T-cells, based on PD-1 and TIGIT co-expression, and a combination with ICI, such as anti-PD-1.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma/inmunología , Linfocitos T/metabolismo , Adulto , Anciano , Línea Celular Tumoral , Humanos , Persona de Mediana Edad
8.
Cell Immunol ; 344: 103961, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31472938

RESUMEN

Merkel cell carcinoma (MCC) is a rare and aggressive cutaneous cancer, which is immunogenic, regardless of the presence of MCPyV (80% of cases). The identification of MCC-specific epitopes recognized by CD8 T cells is crucial to expand the arsenal of immunotherapeutic treatments. Until now, most efforts focused on the identification of virus-specific epitopes, whereas immune responses directed against shared cellular tumor-specific antigens have not been evidenced. In this study, we measured T-cell responses against viral (n = 3) and tumor antigens (n = 47) from TILs derived from 21 MCC tumors. Virus-specific CD8 T-cell responses dominated MCC-specific immune responses, and we identified two new HLA-peptide complexes derived from the LT antigen, located in a region encompassing 3 previously identified epitopes. Finally, we show that MAGE-A3 antigen, frequently expressed by MCC tumors, was recognized by CD8 TILs from a virus-negative MCC tumor and thus could be a target for immunotherapy in this setting.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma de Células de Merkel/inmunología , Neoplasias Cutáneas/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos Virales/inmunología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Epítopos de Linfocito T/inmunología , Femenino , Antígenos HLA/inmunología , Humanos , Masculino , Proteínas de Neoplasias/inmunología
9.
Med Sci (Paris) ; 40(2): 176-185, 2024 Feb.
Artículo en Francés | MEDLINE | ID: mdl-38411426

RESUMEN

The ubiquitin-proteasome system (UPS) is a conserved degradation pathway in eukaryotes, playing a central role in various cellular processes, including maintaining protein homeostasis, regulating the cell cycle and signaling pathways, as well as orchestrating cell survival and death. Proteins targeted for UPS-mediated degradation undergo ubiquitin chain modification before being degraded by 26S proteasomes. Recently, a correlation has emerged between pathogenic proteasome variants and the onset of neurodevelopmental disorders. Termed "neurodevelopmental proteasomopathies", these syndromes are rare and characterized by delayed psychomotor development, behavioral disorders, facial dysmorphia, and multisystemic anomalies. In this review, we examine current knowledge on proteasomal dysfunctions and assess their relevance in the search for biomarkers for the diagnosis and potential treatment of these syndromic proteasomopathies.


Title: Protéasomopathies neurodéveloppementales : une nouvelle classe de maladies du neurodéveloppement causées par une dysfonction du protéasome. Abstract: Le système ubiquitine-protéasome (UPS) est une voie conservée chez les eucaryotes qui permet la dégradation, par les protéasomes, des protéines modifiées par l'ubiquitine. Récemment, une corrélation entre des variants pathogènes de gènes codant le protéasome et l'émergence de nouvelles maladies avec troubles neurodéveloppementaux, dénommés « protéasomopathies neurodéveloppementales ¼, a été mise en évidence. Ces maladies rares se manifestent par des retards psychomoteurs, des troubles du comportement, des dysmorphies faciales et des anomalies multi-systémiques. Dans cette synthèse, nous répertorions les biomarqueurs spécifiques d'une dysfonction protéasomale et nous discutons de leur pertinence pour le diagnostic et les traitements de ces troubles neurodéveloppementaux.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Ubiquitina , Humanos , Citoplasma , Ciclo Celular , División Celular
10.
medRxiv ; 2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38293138

RESUMEN

Neurodevelopmental proteasomopathies represent a distinctive category of neurodevelopmental disorders (NDD) characterized by genetic variations within the 26S proteasome, a protein complex governing eukaryotic cellular protein homeostasis. In our comprehensive study, we identified 23 unique variants in PSMC5 , which encodes the AAA-ATPase proteasome subunit PSMC5/Rpt6, causing syndromic NDD in 38 unrelated individuals. Overexpression of PSMC5 variants altered human hippocampal neuron morphology, while PSMC5 knockdown led to impaired reversal learning in flies and loss of excitatory synapses in rat hippocampal neurons. PSMC5 loss-of-function resulted in abnormal protein aggregation, profoundly impacting innate immune signaling, mitophagy rates, and lipid metabolism in affected individuals. Importantly, targeting key components of the integrated stress response, such as PKR and GCN2 kinases, ameliorated immune dysregulations in cells from affected individuals. These findings significantly advance our understanding of the molecular mechanisms underlying neurodevelopmental proteasomopathies, provide links to research in neurodegenerative diseases, and open up potential therapeutic avenues.

11.
medRxiv ; 2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37398376

RESUMEN

Purpose: De novo variants in CUL3 (Cullin-3 ubiquitin ligase) have been strongly associated with neurodevelopmental disorders (NDDs), but no large case series have been reported so far. Here we aimed to collect sporadic cases carrying rare variants in CUL3, describe the genotype-phenotype correlation, and investigate the underlying pathogenic mechanism. Methods: Genetic data and detailed clinical records were collected via multi-center collaboration. Dysmorphic facial features were analyzed using GestaltMatcher. Variant effects on CUL3 protein stability were assessed using patient-derived T-cells. Results: We assembled a cohort of 35 individuals with heterozygous CUL3 variants presenting a syndromic NDD characterized by intellectual disability with or without autistic features. Of these, 33 have loss-of-function (LoF) and two have missense variants. CUL3 LoF variants in patients may affect protein stability leading to perturbations in protein homeostasis, as evidenced by decreased ubiquitin-protein conjugates in vitro . Specifically, we show that cyclin E1 (CCNE1) and 4E-BP1 (EIF4EBP1), two prominent substrates of CUL3, fail to be targeted for proteasomal degradation in patient-derived cells. Conclusion: Our study further refines the clinical and mutational spectrum of CUL3 -associated NDDs, expands the spectrum of cullin RING E3 ligase-associated neuropsychiatric disorders, and suggests haploinsufficiency via LoF variants is the predominant pathogenic mechanism.

12.
Sci Transl Med ; 15(698): eabo3189, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37256937

RESUMEN

A critical step in preserving protein homeostasis is the recognition, binding, unfolding, and translocation of protein substrates by six AAA-ATPase proteasome subunits (ATPase-associated with various cellular activities) termed PSMC1-6, which are required for degradation of proteins by 26S proteasomes. Here, we identified 15 de novo missense variants in the PSMC3 gene encoding the AAA-ATPase proteasome subunit PSMC3/Rpt5 in 23 unrelated heterozygous patients with an autosomal dominant form of neurodevelopmental delay and intellectual disability. Expression of PSMC3 variants in mouse neuronal cultures led to altered dendrite development, and deletion of the PSMC3 fly ortholog Rpt5 impaired reversal learning capabilities in fruit flies. Structural modeling as well as proteomic and transcriptomic analyses of T cells derived from patients with PSMC3 variants implicated the PSMC3 variants in proteasome dysfunction through disruption of substrate translocation, induction of proteotoxic stress, and alterations in proteins controlling developmental and innate immune programs. The proteostatic perturbations in T cells from patients with PSMC3 variants correlated with a dysregulation in type I interferon (IFN) signaling in these T cells, which could be blocked by inhibition of the intracellular stress sensor protein kinase R (PKR). These results suggest that proteotoxic stress activated PKR in patient-derived T cells, resulting in a type I IFN response. The potential relationship among proteosome dysfunction, type I IFN production, and neurodevelopment suggests new directions in our understanding of pathogenesis in some neurodevelopmental disorders.


Asunto(s)
Interferón Tipo I , Complejo de la Endopetidasa Proteasomal , Animales , Humanos , Ratones , Adenosina Trifosfatasas/genética , Drosophila melanogaster , Expresión Génica , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica
13.
Eur J Immunol ; 40(6): 1786-94, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20217862

RESUMEN

We recently showed that the infusion of tumor infiltrating lymphocytes specific for the MELOE-1 antigen was associated with a prolonged relapse-free survival for HLA-A2(+) melanoma patients who received tumor infiltrating lymphocytes therapy. Here, we characterized the MELOE-1/A2-specific T-cell repertoire in healthy donors and melanoma patients to further support an immunotherapy targeting this epitope. Using tetramer enrichment followed by multicolor staining, we found that MELOE-1-specific T cells were present in the blood of healthy donors and patients at similar frequencies (around 1 in 1x10(5) CD8(+) cells). These cells mainly displayed a naïve phenotype in 4/6 healthy donors and 3/6 patients, whereas high proportions of memory cells were observed in the remaining individuals of both groups. There was a recurrent usage of the Valpha12.1 chain for 17/18 MELOE-1-specific T-cell clones derived from healthy donors or patients, associated with diverse Vbeta chains and V(D)J junctional sequences. All clones derived from melanoma patients (9/9) were reactive against the MELOE-1(36-44) peptide and against HLA-A2(+) melanoma cell lines. This study documents the existence of a large TCR repertoire specific for the MELOE-1/A2 epitope and its capacity to give rise to antitumor CTL that supports the development of immunotherapies targeting this epitope.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia/métodos , Melanoma/inmunología , Proteínas de Neoplasias/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Diferenciación Celular/inmunología , Separación Celular , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/terapia , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/citología , Linfocitos T/citología
14.
Cancer Immunol Immunother ; 60(3): 327-37, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21080167

RESUMEN

CD4(+) T cells contribute importantly to the antitumor T cell response, and thus, long peptides comprising CD4 and CD8 epitopes may be efficient cancer vaccines. We have previously identified an overexpressed antigen in melanoma, MELOE-1, presenting a CD8(+) T cell epitope, MELOE-1(36-44), in the HLA-A*0201 context. A T cell repertoire against this epitope is present in HLA-A*0201+ healthy subjects and melanoma patients and the adjuvant injection of TIL containing MELOE-1 specific CD8(+) T cells to melanoma patients was shown to be beneficial. In this study, we looked for CD4(+) T cell epitopes in the vicinity of the HLA-A*0201 epitope. Stimulation of PBMC from healthy subjects with MELOE-1(26-46) revealed CD4 responses in multiple HLA contexts and by cloning responsive CD4(+) T cells, we identified one HLA-DRß1*1101-restricted and one HLA-DQß1*0603-restricted epitope. We showed that the two epitopes could be efficiently presented to CD4(+) T cells by MELOE-1-loaded dendritic cells but not by MELOE-1+ melanoma cell-lines. Finally, we showed that the long peptide MELOE-1(22-46), containing the two optimal class II epitopes and the HLA-A*0201 epitope, was efficiently processed by DC to stimulate CD4(+) and CD8(+) T cell responses in vitro, making it a potential candidate for melanoma vaccination.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/citología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Antígenos HLA-A/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Melanoma/terapia , Proteínas de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Antígeno HLA-A2 , Humanos , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Melanoma/inmunología , Estándares de Referencia
15.
Cancer Immunol Immunother ; 59(3): 431-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19730858

RESUMEN

We characterized a new melanoma antigen derived from one of the multiple open reading frames (ORFs) of the meloe transcript. The meloe gene is overexpressed in melanomas as compared to other cancer cell lines and normal tissues. The corresponding transcript is rather unusual, in that it does not contain a long unique ORF but multiple short ORFs. We recently characterized a tumor epitope derived from a polypeptide (MELOE-1) encoded by the ORF(1230-1370) and involved in relapse prevention of melanoma patients treated with autologous tumor infiltrating lymphocytes (TIL). Here we show that the ORF(285-404) encodes a polypeptide called MELOE-2 that also generated a HLA-A2 epitope recognized by a melanoma-specific T cell clone derived from the same TIL population from which we derived the MELOE-1-specific T cell clone. We also showed that HLA-A2 melanoma cells were spontaneously recognized by the MELOE-2-specific T cell clone, and we detected the presence of MELOE-2 reactive T cells in another TIL population infused to a patient who remained relapse-free after TIL treatment. These results demonstrate that translation of meloe transcript in melanoma cells can produce at least two immunogenic polypeptides, MELOE-1 and MELOE-2, from two distinct ORFs that could be relevant target for melanoma immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/fisiología , Antígeno HLA-A2/inmunología , Melanoma/genética , Melanoma/inmunología , Proteínas de Neoplasias/inmunología , Proteínas de Neoplasias/fisiología , Sistemas de Lectura Abierta/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Linfocitos T/inmunología , Antígenos de Neoplasias/genética , Secuencia de Bases , Ensayos Clínicos como Asunto , ADN Complementario/química , ADN Complementario/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética
16.
Cancer Immunol Res ; 8(2): 255-267, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31857348

RESUMEN

MicroRNAs (miRNA), small noncoding RNAs that regulate gene expression, exist not only in cells but also in a variety of body fluids. These circulating miRNAs could enable intercellular communication. miRNAs are packaged in membrane-encapsulated vesicles, such as exosomes, or protected by RNA-binding proteins. Here, we report that miRNAs included in human melanoma exosomes regulate the tumor immune response. Using microscopy and flow cytometry, we demonstrate that CD8+ T cells internalize exosomes from different tumor types even if these cells do not internalize vesicles as readily as other immune cells. We explored the function of melanoma-derived exosomes in CD8+ T cells and showed that these exosomes downregulate T-cell responses through decreased T-cell receptor (TCR) signaling and diminished cytokine and granzyme B secretions. The result reduces the cells' cytotoxic activity. Using mimics, we found that miRNAs enriched in exosomes-such as Homo sapiens (hsa)-miR-3187-3p, hsa-miR-498, hsa-miR-122, hsa-miR149, and hsa-miR-181a/b-regulate TCR signaling and TNFα secretion. Our observations suggest that miRNAs in melanoma-derived exosomes aid tumor immune evasion and could be a therapeutic target.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Exosomas/genética , Melanoma/inmunología , MicroARNs/genética , Transducción de Señal , Neoplasias Cutáneas/inmunología , Escape del Tumor , Comunicación Celular , Línea Celular Tumoral , Células Cultivadas , Exosomas/inmunología , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , MicroARNs/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
17.
J Immunother Cancer ; 8(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32001504

RESUMEN

BACKGROUND: Genome editing offers unique perspectives for optimizing the functional properties of T cells for adoptive cell transfer purposes. So far, PDCD1 editing has been successfully tested mainly in chimeric antigen receptor T (CAR-T) cells and human primary T cells. Nonetheless, for patients with solid tumors, the adoptive transfer of effector memory T cells specific for tumor antigens remains a relevant option, and the use of high avidity T cells deficient for programmed cell death-1 (PD-1) expression is susceptible to improve the therapeutic benefit of these treatments. METHODS: Here we used the transfection of CAS9/sgRNA ribonucleoproteic complexes to edit PDCD1 gene in human effector memory CD8+ T cells specific for the melanoma antigen Melan-A. We cloned edited T cell populations and validated PDCD1 editing through sequencing and cytometry in each T cell clone, together with T-cell receptor (TCR) chain's sequencing. We also performed whole transcriptomic analyses on wild-type (WT) and edited T cell clones. Finally, we documented in vitro and in vivo through adoptive transfer in NOD scid gamma (NSG) mice, the antitumor properties of WT and PD-1KO T cell clones, expressing the same TCR. RESULTS: Here we demonstrated the feasibility to edit PDCD1 gene in human effector memory melanoma-specific T lymphocytes. We showed that PD-1 expression was dramatically reduced or totally absent on PDCD1-edited T cell clones. Extensive characterization of a panel of T cell clones expressing the same TCR and exhibiting similar functional avidity demonstrated superior antitumor reactivity against a PD-L1 expressing melanoma cell line. Transcriptomic analysis revealed a downregulation of genes involved in proliferation and DNA replication in PD-1-deficient T cell clones, whereas genes involved in metabolism and cell signaling were upregulated. Finally, we documented the superior ability of PD-1-deficient T cells to significantly delay the growth of a PD-L1 expressing human melanoma tumor in an NSG mouse model. CONCLUSION: The use of such lymphocytes for adoptive cell transfer purposes, associated with other approaches modulating the tumor microenvironment, would be a promising alternative to improve immunotherapy efficacy in solid tumors.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Melanoma/inmunología , Melanoma/terapia , Receptor de Muerte Celular Programada 1/deficiencia , Linfocitos T Citotóxicos/inmunología , Animales , Línea Celular Tumoral , Femenino , Edición Génica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Distribución Aleatoria , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Immunother Cancer ; 8(2)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33188038

RESUMEN

BACKGROUND: Clinical benefit from programmed cell death 1 receptor (PD-1) inhibitors relies on reinvigoration of endogenous antitumor immunity. Nonetheless, robust immunological markers, based on circulating immune cell subsets associated with therapeutic efficacy are yet to be validated. METHODS: We isolated peripheral blood mononuclear cell from three independent cohorts of melanoma and Merkel cell carcinoma patients treated with PD-1 inhibitor, at baseline and longitudinally after therapy. Using multiparameter flow cytometry and cell sorting, we isolated four subsets of CD8+ T cells, based on PD-1 and TIGIT expression profiles. We performed phenotypic characterization, T cell receptor sequencing, targeted transcriptomic analysis and antitumor reactivity assays to thoroughly characterize each of these subsets. RESULTS: We documented that the frequency of circulating PD-1+TIGIT+ (DPOS) CD8+ T-cells after 1 month of anti-PD-1 therapy was associated with clinical response and overall survival. This DPOS T-cell population was enriched in highly activated T-cells, tumor-specific and emerging T-cell clonotypes and T lymphocytes overexpressing CXCR5, a key marker of the CD8 cytotoxic follicular T cell population. Additionally, transcriptomic profiling defined a specific gene signature for this population as well as the overexpression of specific pathways associated with the therapeutic response. CONCLUSIONS: Our results provide a convincing rationale for monitoring this PD-1+TIGIT+ circulating population as an early cellular-based marker of therapeutic response to anti-PD-1 therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Carcinoma de Células de Merkel/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Melanoma/inmunología , Receptor de Muerte Celular Programada 1/biosíntesis , Receptores Inmunológicos/biosíntesis , Linfocitos T CD8-positivos/metabolismo , Carcinoma de Células de Merkel/sangre , Carcinoma de Células de Merkel/tratamiento farmacológico , Humanos , Melanoma/sangre , Melanoma/tratamiento farmacológico , Valor Predictivo de las Pruebas , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/sangre , Receptor de Muerte Celular Programada 1/inmunología , Receptores CXCR5/inmunología , Receptores Inmunológicos/sangre , Receptores Inmunológicos/inmunología , Subgrupos de Linfocitos T/inmunología
19.
J Clin Invest ; 130(11): 6109-6123, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33074246

RESUMEN

T cell exclusion causes resistance to cancer immunotherapies via immune checkpoint blockade (ICB). Myeloid cells contribute to resistance by expressing signal regulatory protein-α (SIRPα), an inhibitory membrane receptor that interacts with ubiquitous receptor CD47 to control macrophage phagocytosis in the tumor microenvironment. Although CD47/SIRPα-targeting drugs have been assessed in preclinical models, the therapeutic benefit of selectively blocking SIRPα, and not SIRPγ/CD47, in humans remains unknown. We report a potent synergy between selective SIRPα blockade and ICB in increasing memory T cell responses and reverting exclusion in syngeneic and orthotopic tumor models. Selective SIRPα blockade stimulated tumor nest T cell recruitment by restoring murine and human macrophage chemokine secretion and increased anti-tumor T cell responses by promoting tumor-antigen crosspresentation by dendritic cells. However, nonselective SIRPα/SIRPγ blockade targeting CD47 impaired human T cell activation, proliferation, and endothelial transmigration. Selective SIRPα inhibition opens an attractive avenue to overcoming ICB resistance in patients with elevated myeloid cell infiltration in solid tumors.


Asunto(s)
Memoria Inmunológica , Inmunoterapia , Neoplasias Mamarias Experimentales/terapia , Proteínas de Neoplasias/inmunología , Receptores Inmunológicos/inmunología , Linfocitos T/inmunología , Animales , Femenino , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/genética , Receptores Inmunológicos/genética , Linfocitos T/patología
20.
Cancer Immunol Immunother ; 58(2): 271-80, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18612636

RESUMEN

Melanoma reactive CTL were obtained by stimulating PBL from a melanoma patient in remission since 1994 following adjuvant TIL immunotherapy, with the autologous melanoma cell line. They were cloned by limiting dilution. One CTL clone recognized melanoma cell lines expressing tyrosinase and the B*4002 molecule, either spontaneously or upon transfection. We demonstrated that this clone recognizes the tyrosinase-derived nonapeptide 316-324 (ADVEFCLSL) and the overlapping decapeptide 315-324 (SADVEFCLSL). We derived two distinct additional specific CTL clones from this same patient that were also reactive against B*4002 melanoma cell lines, suggesting a relative diversity of this specific repertoire in this patient. Stimulating PBMC derived from four additional B*4002 melanoma patients with the tyrosinase 316-324 nonapeptide induced the growth of specific cells for two of the patients, demonstrating the immunogenicity of this new epitope. Our data show that this nonapeptide is a new tool that could be used to generate melanoma-specific T cells for adoptive immunotherapy or serve as a peptide vaccine for HLA-B*4002 melanoma patients.


Asunto(s)
Epítopos/inmunología , Antígenos HLA-B/inmunología , Melanoma/inmunología , Monofenol Monooxigenasa/inmunología , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , ADN Complementario/genética , Epítopos/genética , Galanina/metabolismo , Antígenos HLA-B/genética , Humanos , Leucocitos Mononucleares/metabolismo , Ratones , Datos de Secuencia Molecular , Monofenol Monooxigenasa/química , Monofenol Monooxigenasa/genética , Neuropéptido Y/análogos & derivados , Neuropéptido Y/metabolismo , Fragmentos de Péptidos/metabolismo , Péptidos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA