Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(6): 735-745, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34017124

RESUMEN

Regulatory T (Treg) cells are a barrier for tumor immunity and a target for immunotherapy. Using single-cell transcriptomics, we found that CD4+ T cells infiltrating primary and metastatic colorectal cancer and non-small-cell lung cancer are highly enriched for two subsets of comparable size and suppressor function comprising forkhead box protein P3+ Treg and eomesodermin homolog (EOMES)+ type 1 regulatory T (Tr1)-like cells also expressing granzyme K and chitinase-3-like protein 2. EOMES+ Tr1-like cells, but not Treg cells, were clonally related to effector T cells and were clonally expanded in primary and metastatic tumors, which is consistent with their proliferation and differentiation in situ. Using chitinase-3-like protein 2 as a subset signature, we found that the EOMES+ Tr1-like subset correlates with disease progression but is also associated with response to programmed cell death protein 1-targeted immunotherapy. Collectively, these findings highlight the heterogeneity of Treg cells that accumulate in primary tumors and metastases and identify a new prospective target for cancer immunotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Hematopoyesis Clonal/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/secundario , Carcinoma de Pulmón de Células no Pequeñas/terapia , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/genética , Quimioterapia Adyuvante/métodos , Quitinasas/metabolismo , Colectomía , Colon/patología , Colon/cirugía , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Resistencia a Antineoplásicos/inmunología , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/inmunología , Granzimas/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , RNA-Seq , Análisis de la Célula Individual , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/metabolismo
2.
Immunity ; 45(5): 1135-1147, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851914

RESUMEN

Tumor-infiltrating regulatory T lymphocytes (Treg) can suppress effector T cells specific for tumor antigens. Deeper molecular definitions of tumor-infiltrating-lymphocytes could thus offer therapeutic opportunities. Transcriptomes of T helper 1 (Th1), Th17, and Treg cells infiltrating colorectal or non-small-cell lung cancers were compared to transcriptomes of the same subsets from normal tissues and validated at the single-cell level. We found that tumor-infiltrating Treg cells were highly suppressive, upregulated several immune-checkpoints, and expressed on the cell surfaces specific signature molecules such as interleukin-1 receptor 2 (IL1R2), programmed death (PD)-1 Ligand1, PD-1 Ligand2, and CCR8 chemokine, which were not previously described on Treg cells. Remarkably, high expression in whole-tumor samples of Treg cell signature genes, such as LAYN, MAGEH1, or CCR8, correlated with poor prognosis. Our findings provide insights into the molecular identity and functions of human tumor-infiltrating Treg cells and define potential targets for tumor immunotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Pulmonares/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Reguladores/inmunología , Anciano , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Separación Celular , Neoplasias Colorrectales/mortalidad , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Pronóstico , Transcriptoma
3.
Eur J Immunol ; 51(12): 3243-3246, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34528258

RESUMEN

Ex vivo gene expression and miRNA profiling of Eomes+ Tr1-like cells suggested that they represent a differentiation stage that is intermediate between Th1-cells and cytotoxic CD4+ T-cells. Several microRNAs were downregulated in Eomes+ Tr1-like cells that might inhibit Tr1-cell differentiation. In particular, miR-92a targeted Eomes, while miR-125a inhibited IFN-g and IL-10R expression.


Asunto(s)
Perfilación de la Expresión Génica , MicroARNs/inmunología , Receptores de Interleucina-10/inmunología , Proteínas de Dominio T Box/inmunología , Células TH1/inmunología , Humanos
4.
Nat Chem Biol ; 15(11): 1077-1084, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31591562

RESUMEN

Cereblon (CRBN) is a primary target of thalidomide and mediates its multiple pharmacological activities, including teratogenic and antimyeloma activities. CRBN functions as a substrate receptor of the E3 ubiquitin ligase CRL4, whose substrate specificity is modulated by thalidomide and its analogs. Although a number of CRL4CRBN substrates have recently been identified, the substrate involved in thalidomide teratogenicity is unclear. Here we show that p63 isoforms are thalidomide-dependent CRL4CRBN neosubstrates that are responsible, at least in part, for its teratogenic effects. The p53 family member p63 is associated with multiple developmental processes. ∆Np63α is essential for limb development, while TAp63α is important for cochlea development and hearing. Using a zebrafish model, we demonstrate that thalidomide exerts its teratogenic effects on pectoral fins and otic vesicles by inducing the degradation of ∆Np63α and TAp63α, respectively. These results may contribute to the invention of new thalidomide analogs lacking teratogenic activity.


Asunto(s)
Proteínas de la Membrana/metabolismo , Teratógenos/toxicidad , Talidomida/toxicidad , Células HEK293 , Humanos , Especificidad por Sustrato
5.
Eur J Immunol ; 49(1): 96-111, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30431161

RESUMEN

Whether human IL-10-producing regulatory T cells ("Tr1") represent a distinct differentiation lineage or an unstable activation stage remains a key unsolved issue. Here, we report that Eomesodermin (Eomes) acted as a lineage-defining transcription factor in human IFN-γ/IL-10 coproducing Tr1-like cells. In vivo occurring Tr1-like cells expressed Eomes, and were clearly distinct from all other CD4+ T-cell subsets, including conventional cytotoxic CD4+ T cells. They expressed Granzyme (Gzm) K, but had lost CD40L and IL-7R expression. Eomes antagonized the Th17 fate, and directly controlled IFN-γ and GzmK expression. However, Eomes binding to the IL-10 promoter was not detectable in human CD4+ T cells, presumably because critical Tbox binding sites of the mouse were not conserved. A precommitment to a Tr1-like fate, i.e. concominant induction of Eomes, GzmK, and IFN-γ, was promoted by IL-4 and IL-12-secreting myeloid dendritic cells. Consistently, Th1 effector memory cells contained precommitted Eomes+ GzmK+ T cells. Stimulation with T-cell receptor (TCR) agonists and IL-27 promoted the generation of Tr1-like effector cells by inducing switching from CD40L to IL-10. Importantly, CD4+ Eomes+ T-cell subsets were present in lymphoid and nonlymphoid tissues, and their frequencies varied systemically in patients with inflammatory bowel disease and graft-versus-host disease. We propose that Eomes+ Tr1-like cells are effector cells of a unique GzmK-expressing CD4+ T-cell subset.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Regulación de la Expresión Génica , Granzimas/metabolismo , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Ratones , Proteínas de Dominio T Box/genética
6.
Cereb Cortex ; 28(5): 1846-1856, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29351579

RESUMEN

During differentiation, neurons progressively restrict their fate repressing the expression of specific genes. Here we describe the involvement in such developmental steps of the methyl-CpG binding protein 2 (MeCP2), an epigenetic factor that participates to chromatin folding and transcriptional regulation. We previously reported that, due to transcriptional impairments, the maturation of Mecp2 null neurons is delayed. To evaluate whether this could stem from altered progenitors proliferation and differentiation, we investigated whether lack of Mecp2 affects these features both in vitro and in vivo. We show that in Mecp2 null embryonic cortexes the expression of genes defining the identity of proliferating neuroprogenitors is enriched and that their permanence in the G1 phase is prolonged. Moreover, the number of cells transitioning from a stage of maturation to a more mature one is increased in Mecp2 null embryonic cortices, in line with the central role of G1 for cell identity refinement. We thus suggest that, possibly due to the lack of proper transcriptional control normally exerted by Mecp2, fate refinement is impaired in developing null cells. We propose that the maturation delay affecting the developing Mecp2 null cortex originates, at least in part, from deranged mechanisms of cell fate refinement.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Neuronas/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina , Células Cultivadas , Ciclina D1/metabolismo , Desoxiuridina/análogos & derivados , Desoxiuridina/toxicidad , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Embarazo , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero
7.
J Allergy Clin Immunol ; 140(3): 797-808, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28237728

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) that is caused by autoreactive T cells and associated with viral infections. However, the phenotype of pathogenic T cells in peripheral blood remains to be defined, and how viruses promote MS is debated. OBJECTIVE: We aimed to identify and characterize potentially pathogenic autoreactive T cells, as well as protective antiviral T cells, in patients with MS. METHODS: We analyzed CD4+ helper T-cell subsets from peripheral blood or cerebrospinal fluid for cytokine production, gene expression, plasticity, homing potentials, and their reactivity to self-antigens and viral antigens in healthy subjects and patients with MS. Moreover, we monitored their frequencies in untreated and fingolimod- or natalizumab-treated patients with MS. RESULTS: TH1/TH17 central memory (TH1/TH17CM) cells were selectively increased in peripheral blood of patients with relapsing-remitting MS with a high disease score. TH1/TH17CM cells were closely related to conventional TH17 cells but had more pathogenic features. In particular, they could shuttle between lymph nodes and the CNS and produced encephalitogenic cytokines. The cerebrospinal fluid of patients with active MS was enriched for CXCL10 and contained mainly CXCR3-expressing TH1 and TH1/TH17 subsets. However, while TH1 cells responded consistently to viruses, TH1/TH17CM cells reacted strongly with John Cunningham virus in healthy subjects but responded instead to myelin-derived self-antigens in patients with MS. Fingolimod and natalizumab therapies efficiently targeted autoreactive TH1/TH17CM cells but also blocked virus-specific TH1 cells. CONCLUSIONS: We propose that autoreactive TH1/TH17CM cells expand in patients with MS and promote relapses after bystander recruitment to the CNS, whereas TH1 cells perform immune surveillance. Thus the selective targeting of TH1/TH17 cells could inhibit relapses without causing John Cunningham virus-dependent progressive multifocal encephalomyelitis.


Asunto(s)
Antígenos Virales/inmunología , Autoantígenos/inmunología , Virus JC/inmunología , Esclerosis Múltiple/inmunología , Células TH1/inmunología , Células Th17/inmunología , Adulto , Citocinas/líquido cefalorraquídeo , Citocinas/inmunología , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Expresión Génica , Humanos , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/líquido cefalorraquídeo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Natalizumab/uso terapéutico
8.
J Immunol ; 193(5): 2196-206, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063866

RESUMEN

We identified two mast cell subsets characterized by the differential expression of surface CD25 (IL-2Rα) and by different abilities to produce cytokines and to proliferate, both in vitro and in vivo. CD25 can be expressed on the surface of immune cells in the absence of the other chains of the IL-2R, which are indispensable for IL-2 signaling. We show that functional differences between the two mast cell populations were dependent on CD25 itself, which directly modulated proliferation and cytokine responses. These effects were completely independent from IL-2 or the expression of the other chains of the high-affinity IL-2R, indicating an autonomous and previously unappreciated role for CD25 in regulating cell functions. Cells genetically ablated for CD25 completely recapitulated the CD25-negative phenotype and never acquired the properties characteristic of CD25-positive mast cells. Finally, adoptive transfer experiments in the mouse demonstrated a different impact of these populations in models of anaphylaxis and contact sensitivity. Our findings indicate a general role for CD25 in contexts where IL-2 signaling is not involved, and may have important implications for all mast cell-related diseases, as well as in all cell types expressing CD25 independently of its IL-2-related functions.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Interleucina-2/inmunología , Mastocitos/inmunología , Transducción de Señal/inmunología , Células 3T3 , Animales , Regulación de la Expresión Génica/genética , Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/genética , Mastocitos/citología , Ratones , Ratones Noqueados , Transducción de Señal/genética
9.
Nucleic Acids Res ; 41(6): 3600-18, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23393190

RESUMEN

Co-option of cis-regulatory modules has been suggested as a mechanism for the evolution of expression sites during development. However, the extent and mechanisms involved in mobilization of cis-regulatory modules remains elusive. To trace the history of non-coding elements, which may represent candidate ancestral cis-regulatory modules affirmed during chordate evolution, we have searched for conserved elements in tunicate and vertebrate (Olfactores) genomes. We identified, for the first time, 183 non-coding sequences that are highly conserved between the two groups. Our results show that all but one element are conserved in non-syntenic regions between vertebrate and tunicate genomes, while being syntenic among vertebrates. Nevertheless, in all the groups, they are significantly associated with transcription factors showing specific functions fundamental to animal development, such as multicellular organism development and sequence-specific DNA binding. The majority of these regions map onto ultraconserved elements and we demonstrate that they can act as functional enhancers within the organism of origin, as well as in cross-transgenesis experiments, and that they are transcribed in extant species of Olfactores. We refer to the elements as 'Olfactores conserved non-coding elements'.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Urocordados/genética , Vertebrados/genética , Animales , Secuencia de Bases , Secuencia Conservada , Perros , Peces/genética , Redes Reguladoras de Genes , Genes Homeobox , Sitios Genéticos , Genoma , Humanos , Mamíferos/genética , Ratones , Sintenía , Transcripción Genética
10.
Nat Commun ; 15(1): 2498, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509063

RESUMEN

T cell-based immunotherapies have exhibited promising outcomes in tumor control; however, their efficacy is limited in immune-excluded tumors. Cancer-associated fibroblasts (CAFs) play a pivotal role in shaping the tumor microenvironment and modulating immune infiltration. Despite the identification of distinct CAF subtypes using single-cell RNA-sequencing (scRNA-seq), their functional impact on hindering T-cell infiltration remains unclear, particularly in soft-tissue sarcomas (STS) characterized by low response rates to T cell-based therapies. In this study, we characterize the STS microenvironment using murine models (in female mice) with distinct immune composition by scRNA-seq, and identify a subset of CAFs we termed glycolytic cancer-associated fibroblasts (glyCAF). GlyCAF rely on GLUT1-dependent expression of CXCL16 to impede cytotoxic T-cell infiltration into the tumor parenchyma. Targeting glycolysis decreases T-cell restrictive glyCAF accumulation at the tumor margin, thereby enhancing T-cell infiltration and augmenting the efficacy of chemotherapy. These findings highlight avenues for combinatorial therapeutic interventions in sarcomas and possibly other solid tumors. Further investigations and clinical trials are needed to validate these potential strategies and translate them into clinical practice.


Asunto(s)
Fibroblastos Asociados al Cáncer , Sarcoma , Neoplasias de los Tejidos Blandos , Femenino , Animales , Ratones , Resistencia a Antineoplásicos , Sarcoma/tratamiento farmacológico , Sarcoma/genética , Linfocitos T Citotóxicos , Microambiente Tumoral , Fibroblastos
11.
Nat Commun ; 14(1): 4703, 2023 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-37543621

RESUMEN

TGFß signaling is associated with non-response to immune checkpoint blockade in patients with advanced cancers, particularly in the immune-excluded phenotype. While previous work demonstrates that converting tumors from excluded to inflamed phenotypes requires attenuation of PD-L1 and TGFß signaling, the underlying cellular mechanisms remain unclear. Here, we show that TGFß and PD-L1 restrain intratumoral stem cell-like CD8 T cell (TSCL) expansion and replacement of progenitor-exhausted and dysfunctional CD8 T cells with non-exhausted T effector cells in the EMT6 tumor model in female mice. Upon combined TGFß/PD-L1 blockade IFNγhi CD8 T effector cells show enhanced motility and accumulate in the tumor. Ensuing IFNγ signaling transforms myeloid, stromal, and tumor niches to yield an immune-supportive ecosystem. Blocking IFNγ abolishes the anti-PD-L1/anti-TGFß therapy efficacy. Our data suggest that TGFß works with PD-L1 to prevent TSCL expansion and replacement of exhausted CD8 T cells, thereby maintaining the T cell compartment in a dysfunctional state.


Asunto(s)
Antígeno B7-H1 , Neoplasias de la Mama , Linfocitos T CD8-positivos , Inhibidores de Puntos de Control Inmunológico , Factor de Crecimiento Transformador beta , Femenino , Animales , Ratones , Diferenciación Celular , Linfocitos T CD8-positivos/inmunología , Células Madre , Antígeno B7-H1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Interferón gamma/inmunología , Agotamiento de Células T , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Endogámicos BALB C , Línea Celular Tumoral , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , RNA-Seq
12.
J Cell Sci ; 123(Pt 14): 2423-33, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20571051

RESUMEN

Tight control of p63 protein levels must be achieved under differentiation or apoptotic conditions. Here, we describe a new regulatory pathway for the DeltaNp63alpha protein. We found that MDM2 binds DeltaNp63alpha in the nucleus promoting its translocation to the cytoplasm. The MDM2 nuclear localization signal is required for DeltaNp63alpha nuclear export and subsequent degradation, whereas the MDM2 ring-finger domain is dispensable. Once exported to the cytoplasm by MDM2, p63 is targeted for degradation by the Fbw7 E3-ubiquitin ligase. Efficient degradation of DeltaNp63alpha by Fbw7 (also known as FBXW7) requires GSK3 kinase activity. By deletion and point mutations analysis we have identified a phosphodegron located in the alpha and beta tail of p63 that is required for degradation. Furthermore, we show that MDM2 or Fbw7 depletion inhibits degradation of endogenous DeltaNp63alpha in cells exposed to UV irradiation, adriamycin and upon keratinocyte differentiation. Our findings suggest that following DNA damage and cellular differentiation MDM2 and Fbw7 can cooperate to regulate the levels of the pro-proliferative DeltaNp63alpha protein.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas F-Box/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Transporte Activo de Núcleo Celular/efectos de la radiación , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Daño del ADN/genética , Doxorrubicina/farmacología , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Ratones , Mutación/genética , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , ARN Interferente Pequeño/genética , Transactivadores/genética , Factores de Transcripción , Activación Transcripcional/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Rayos Ultravioleta/efectos adversos
13.
Nucleic Acids Res ; 38(Database issue): D814-21, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19906694

RESUMEN

PRGdb is a web accessible open-source (http://www.prgdb.org) database that represents the first bioinformatic resource providing a comprehensive overview of resistance genes (R-genes) in plants. PRGdb holds more than 16,000 known and putative R-genes belonging to 192 plant species challenged by 115 different pathogens and linked with useful biological information. The complete database includes a set of 73 manually curated reference R-genes, 6308 putative R-genes collected from NCBI and 10463 computationally predicted putative R-genes. Thanks to a user-friendly interface, data can be examined using different query tools. A home-made prediction pipeline called Disease Resistance Analysis and Gene Orthology (DRAGO), based on reference R-gene sequence data, was developed to search for plant resistance genes in public datasets such as Unigene and Genbank. New putative R-gene classes containing unknown domain combinations were discovered and characterized. The development of the PRG platform represents an important starting point to conduct various experimental tasks. The inferred cross-link between genomic and phenotypic information allows access to a large body of information to find answers to several biological questions. The database structure also permits easy integration with other data types and opens up prospects for future implementations.


Asunto(s)
Biología Computacional/métodos , Bases de Datos Genéticas , Bases de Datos de Ácidos Nucleicos , Perfilación de la Expresión Génica , Genes de Plantas/genética , Genoma de Planta , Enfermedades de las Plantas/genética , Biología Computacional/tendencias , Bases de Datos de Proteínas , Almacenamiento y Recuperación de la Información/métodos , Internet , Estrés Oxidativo , Plantas/metabolismo , Estructura Terciaria de Proteína , Programas Informáticos , Interfaz Usuario-Computador
14.
Cell Rep ; 39(12): 110977, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35732118

RESUMEN

The standard of care is unsuccessful to treat recurrent and aggressive soft-tissue sarcomas. Interventions aimed at targeting components of the tumor microenvironment have shown promise for many solid tumors yet have been only marginally tested for sarcoma, partly because knowledge of the sarcoma microenvironment composition is limited. We employ single-cell RNA sequencing to characterize the immune composition of an undifferentiated pleiomorphic sarcoma mouse model, showing that macrophages in the sarcoma mass exhibit distinct activation states. Sarcoma cells use the pleiotropic cytokine macrophage migration inhibitory factor (MIF) to interact with macrophages expressing the CD74 receptor to switch macrophages' activation state and pro-tumorigenic potential. Blocking the expression of MIF in sarcoma cells favors the accumulation of macrophages with inflammatory and antigen-presenting profiles, hence reducing tumor growth. These data may pave the way for testing new therapies aimed at re-shaping the sarcoma microenvironment, in combination with the standard of care.


Asunto(s)
Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Sarcoma , Neoplasias de los Tejidos Blandos , Animales , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , RNA-Seq , Sarcoma/genética , Microambiente Tumoral
15.
Cell Death Differ ; 29(3): 614-626, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34845371

RESUMEN

High Grade Serous Ovarian cancer (HGSOC) is a major unmet need in oncology, due to its precocious dissemination and the lack of meaningful human models for the investigation of disease pathogenesis in a patient-specific manner. To overcome this roadblock, we present a new method to isolate and grow single cells directly from patients' metastatic ascites, establishing the conditions for propagating them as 3D cultures that we refer to as single cell-derived metastatic ovarian cancer spheroids (sMOCS). By single cell RNA sequencing (scRNAseq) we define the cellular composition of metastatic ascites and trace its propagation in 2D and 3D culture paradigms, finding that sMOCS retain and amplify key subpopulations from the original patients' samples and recapitulate features of the original metastasis that do not emerge from classical 2D culture, including retention of individual patients' specificities. By enabling the enrichment of uniquely informative cell subpopulations from HGSOC metastasis and the clonal interrogation of their diversity at the functional and molecular level, this method provides a powerful instrument for precision oncology in ovarian cancer.


Asunto(s)
Ascitis , Neoplasias Ováricas , Ascitis/genética , Ascitis/patología , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Ováricas/patología , Medicina de Precisión , Esferoides Celulares/patología
16.
Nat Commun ; 13(1): 7243, 2022 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-36433954

RESUMEN

Exonic circular RNAs (circRNAs) produce predominantly non-coding RNA species that have been recently profiled in many tumors. However, their functional contribution to cancer progression is still poorly understood. Here, we identify the circRNAs expressed in soft tissue sarcoma cells and explore how the circRNAs regulate sarcoma growth in vivo. We show that circCsnk1g3 and circAnkib1 promote tumor growth by shaping a pro-tumorigenic microenvironment, possibly due to their capabilities to regulate tumor-promoting elements extrinsic to the tumor cells. Accordingly, circCsnk1g3 and circAnkib1 can control the expression of interferon-related genes and pro-inflammatory factors in the sarcoma cells, thus directing immune cell recruitment into the tumor mass, and hence their activation. Mechanistically, circRNAs may repress pro-inflammatory elements by buffering activation of the pathways mediated by RIG-I, the cytosolic viral RNA sensor. The current findings suggest that the targeting of specific circRNAs could augment the efficacy of tumor and immune response to mainstay therapies.


Asunto(s)
Carcinogénesis , Interferones , ARN Circular , Sarcoma , Neoplasias de los Tejidos Blandos , Microambiente Tumoral , Humanos , Carcinogénesis/genética , Carcinogénesis/inmunología , Interferones/genética , Interferones/inmunología , ARN Circular/genética , ARN Circular/inmunología , Sarcoma/genética , Sarcoma/inmunología , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Quinasa de la Caseína I/genética , Quinasa de la Caseína I/inmunología
17.
Eur Heart J ; 31(22): 2765-73, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20534597

RESUMEN

AIMS: Circulating microRNAs (miRNAs) may represent a novel class of biomarkers; therefore, we examined whether acute myocardial infarction (MI) modulates miRNAs plasma levels in humans and mice. METHODS AND RESULTS: Healthy donors (n = 17) and patients (n = 33) with acute ST-segment elevation MI (STEMI) were evaluated. In one cohort (n = 25), the first plasma sample was obtained 517 ± 309 min after the onset of MI symptoms and after coronary reperfusion with percutaneous coronary intervention (PCI); miR-1, -133a, -133b, and -499-5p were ~15- to 140-fold control, whereas miR-122 and -375 were ~87-90% lower than control; 5 days later, miR-1, -133a, -133b, -499-5p, and -375 were back to baseline, whereas miR-122 remained lower than control through Day 30. In additional patients (n = 8; four treated with thrombolysis and four with PCI), miRNAs and troponin I (TnI) were quantified simultaneously starting 156 ± 72 min after the onset of symptoms and at different times thereafter. Peak miR-1, -133a, and -133b expression and TnI level occurred at a similar time, whereas miR-499-5p exhibited a slower time course. In mice, miRNAs plasma levels and TnI were measured 15 min after coronary ligation and at different times thereafter. The behaviour of miR-1, -133a, -133b, and -499-5p was similar to STEMI patients; further, reciprocal changes in the expression levels of these miRNAs were found in cardiac tissue 3-6 h after coronary ligation. In contrast, miR-122 and -375 exhibited minor changes and no significant modulation. In mice with acute hind-limb ischaemia, there was no increase in the plasma level of the above miRNAs. CONCLUSION: Acute MI up-regulated miR-1, -133a, -133b, and -499-5p plasma levels, both in humans and mice, whereas miR-122 and -375 were lower than control only in STEMI patients. These miRNAs represent novel biomarkers of cardiac damage.


Asunto(s)
MicroARNs/metabolismo , Infarto del Miocardio/diagnóstico , Adulto , Anciano , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Troponina I/metabolismo
18.
EMBO Mol Med ; 13(4): e12433, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33665914

RESUMEN

MECP2 mutations cause Rett syndrome (RTT), a severe and progressive neurodevelopmental disorder mainly affecting females. Although RTT patients exhibit delayed onset of symptoms, several evidences demonstrate that MeCP2 deficiency alters early development of the brain. Indeed, during early maturation, Mecp2 null cortical neurons display widespread transcriptional changes, reduced activity, and defective morphology. It has been proposed that during brain development these elements are linked in a feed-forward cycle where neuronal activity drives transcriptional and morphological changes that further increase network maturity. We hypothesized that the enhancement of neuronal activity during early maturation might prevent the onset of RTT-typical molecular and cellular phenotypes. Accordingly, we show that the enhancement of excitability, obtained by adding to neuronal cultures Ampakine CX546, rescues transcription of several genes, neuronal morphology, and responsiveness to stimuli. Greater effects are achieved in response to earlier treatments. In vivo, short and early administration of CX546 to Mecp2 null mice prolongs lifespan, delays the disease progression, and rescues motor abilities and spatial memory, thus confirming the value for RTT of an early restoration of neuronal activity.


Asunto(s)
Proteína 2 de Unión a Metil-CpG , Síndrome de Rett , Animales , Encéfalo/metabolismo , Femenino , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Neuronas/metabolismo , Fenotipo , Síndrome de Rett/genética
19.
Science ; 372(6542)2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33958447

RESUMEN

Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.


Asunto(s)
Atlas como Asunto , Cuerpo Estriado/citología , Cuerpo Estriado/embriología , Neurogénesis/genética , ARN Largo no Codificante/genética , Análisis de la Célula Individual , Factores de Transcripción/genética , Feto , Neuronas GABAérgicas/metabolismo , Humanos , RNA-Seq , Transcripción Genética
20.
Nat Commun ; 12(1): 2340, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879786

RESUMEN

Cancer is characterized by pervasive epigenetic alterations with enhancer dysfunction orchestrating the aberrant cancer transcriptional programs and transcriptional dependencies. Here, we epigenetically characterize human colorectal cancer (CRC) using de novo chromatin state discovery on a library of different patient-derived organoids. By exploring this resource, we unveil a tumor-specific deregulated enhancerome that is cancer cell-intrinsic and independent of interpatient heterogeneity. We show that the transcriptional coactivators YAP/TAZ act as key regulators of the conserved CRC gained enhancers. The same YAP/TAZ-bound enhancers display active chromatin profiles across diverse human tumors, highlighting a pan-cancer epigenetic rewiring which at single-cell level distinguishes malignant from normal cell populations. YAP/TAZ inhibition in established tumor organoids causes extensive cell death unveiling their essential role in tumor maintenance. This work indicates a common layer of YAP/TAZ-fueled enhancer reprogramming that is key for the cancer cell state and can be exploited for the development of improved therapeutic avenues.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias Colorrectales/genética , Elementos de Facilitación Genéticos , Epigénesis Genética , Transactivadores/genética , Factores de Transcripción/genética , Regulación Neoplásica de la Expresión Génica , Código de Histonas , Humanos , Modelos Genéticos , Organoides/metabolismo , RNA-Seq , Análisis de la Célula Individual , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Células Tumorales Cultivadas , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA